The 2011 Bio-IT World Conference & Expo – On the way to Personalized Predictive Preventative Participatory Medicine

I introduced the PPPPM concept a year ago in a blog post Harnessing the engines of finance and commerce for life-extension.   I wrote that post just after having attended the 2010 version of the 2010 Bio-IT World Conference & Expo.  I speculated that PPPPM will create a basic shift that profoundly affects our health and longevity.  In that blog entry I characterized PPPPM this way: 

“1. The objective of PPPPM is not so much to cure diseases as it is to detect and predict disease susceptibilities before a disease starts or at very early stages of disease progression and initiate personalized interventions to prevent the progression of the disease before it becomes symptomatic or does damage.

 2. PPPPM is participatory in the sense that the collaborative participation of large numbers of health research institutions and care agencies is involved in doing the research and creating the infrastructure to make it workable. It involves a tight and continuing linkup loop of researchers, practitioners, patients and healthy people who want to stay healthy.

 3. The predictions of PPPPM are based on the identification of sets of biomarkers that are predictive of disease susceptibilities and stages of disease progression for particular diseases. The biomarkers can consist of known gene mutations, SNPs, copy number variations and the like, and other “omics” markers (proteomic, transcriptomic, metabolomic, epigenomic etc.) as well as the results of all kinds of existing clinical tests and clinical data.

 4. The biomarkers will be arrived at through massive correlation analyses and pattern matching between public data bases of the kinds of data involved and association studies of many different kinds. See the blog entry Genome-wide association studies for examples. The biomarkers will be continuously refined through feedback from personal histories, of course with numerous layers of personal privacy protection.

 5. Identifying PPPPM biomarkers will proceed one disease at a time, the challenges including creation of massive public data bases of “omics” information and performing multivariate association studies. It is a task that requires mobilization of incredible networked computer power and human analytics.

 6. Identifications of disease-prevention interventions including drug candidates will proceed along the same lines using the same kinds of tools and analytics, where interventions will be determined on the basis of the known biomarker patterns as well as individual patient or well-persons’ patterns of “omic” markers.

 7. Fully implementing PPPPM will require genetic, genomic and other “omic” profiling on the part of increasing numbers of people, something that should become commonplace in 10-15 years.”

I was excited when writing this a year ago because I saw PPPPM as an important emerging paradigm for medicine.  I am even more excited now because a) I see the emerging PPPM paradigm as being much more overarching.  It applies to universal health, wellbeing and longevity, not just to the body-repair business that most medicine is engaged in today, b) because the eventual components of PPPPM are becoming better identified, c) because concrete things are happening on a significant scale that are helping PPPPM become reality, and c) because the pace of these developments is accelerating. Things are happening faster than I thought they would.

This year’s Bio-IT World Conference & Expo was a hybrid event of a major professional conference with six simultaneous tracks of presentations, 43 poster presentations together with an industry trade show featuring some 111 exhibitors.  The title of the conference suggests that it is about where information technology meets life sciences and biological research and the front of the conference program suggests that it is about “Enabling Technology, Leveraging Data, Transforming Medicine,” but these only begin to suggest what the conference is ultimately about.  I believe that taken together the conference is about developments that will lead to an expanded vision of PPPPM.

Here is my take on that expanded version of the PPPPM vision:

 1)    Suppose that we could over time make virtually all research and clinical information and data relating to every disease and every health-and aging related biological pathway electronically available.  I am talking here about all the world’s published scientific literature, the laboratory data on which those publications are based, all information related to drugs and the health effects of lifestyle patterns, all genomic, epigenomic and other “omics” databases, diets and nutraceuticals, all information about every known disease and all patient history records.  We already have a number of piecewise starts in assembling such information although these initiatives are not integrated.  And some important kinds of information are not yet being systematically gathered.  For example, there are few systematic databases characterizing the long-term outcomes of drug, surgical, and other medical interventions.  And we are only now beginning to appreciate the importance of other important kinds of information such as the genetic variability among individuals and the epigenetic and epigenomic effects of environmental and dietary patterns on specific individuals. 

2)    Suppose that we could multidimensionality structure and organize that information and data in flexible ways into databases that that reflects our evolving best scientific models of health and disease states – such as according to molecular pathways and gene activation networks as well as cell and organ systems impacts.  It is important that the data structuring and organization be flexible and allow constant modification as our state of knowledge expands. 

3)    Simultaneous with this, suppose we develop increasingly sophisticated computer models of healthy body states, aging  and disease states – models that relate the mountains of general and individual information to human health outcomes, aging and specific diseases. Identification of disease biomarkers becomes just part of the modeling effort that is required.

4)    Then, so the hypothesis goes, we could do amazing things like;

a.      generate individualized health and disease susceptibility predictions,

b.      identify individualized lifestyle regimens and interventions for optimizing health and assuring longevity,

c.      create a situation where health maintenance is the main thing to be concerned with in life and where attention to diseases become the exception rather than the rule,

d.     vastly speed up the process of drug discovery and approval,

e.     customize medical and drug treatments to the specifics of an individual when a disease does occur, and

f.       even allow drugs to be custom-compounded so that a drug is tailored to meet the particular needs of an individual in a particular condition at a particular moment.

 Strong elements of 1), 2) and 3) above already exist and new more-comprehensive initiatives are getting underway.  These components of what eventually will be PPPPM are being created not as a result of some master plan, but as a result of a rich variety of bottom-up initiatives using a variety of organizational and technical approaches. These approaches are allowing the eventual form of PPPPM to be shaped by an evolutionary process that can work in the presence of current social, business and economic constraints rather than by any current blueprint of what we might think is an ideal approach.   We now have some partial models of PPPPM in actual operation and we have identified a number of important guiding concepts and trends. 

 In the rest of this blog entry I focus on the emerging concepts and trends related to PPPPM.   These are concepts and trends expressed repeatedly in the technical presentations and vendor exhibits at the 2011 Bio-IT World Conference & Expo.  I expect to describe one or more concrete examples of PPPPM-like initiatives in subsequent blog entries.  

Emerging concepts and trends pertaining to PPPPM

The big pharma crunch 

Major pharmaceutical companies have been sitting on large cash reserves but are facing into a crunch having to do with declining productivity of traditional approaches to new drug discovery and development.  While more and more money is being spent on drug discovery and R&D and more and more drugs are in the pipeline, fewer and fewer new drugs are making it through the development process and the FDA approval pipeline.  At the same time, more and more of the traditional money-making blockbuster drugs are going off of patent and becoming low-margin generics.  Written back in 2009 in an opinion piece Crunch time for pharma:  “To illustrate my concerns, let’s look at the treatment of heart disease. Many important cardiovascular drugs have been invented: statins, ACE inhibitors, beta blockers, fibrinolytics. But in the last 10 years, few of significance have emerged, even though the pharmaceutical industry has spent unprecedented amounts of money on research and development: in each year of that decade, Pfizer spent about $6 billion, Eli Lilly $3bn, and GlaxoSmithKline $2.5bn.”  “I believe that there is a real risk that the big pharma industry might collapse.”  The 2010 Pharma R&D Annual Review said “Outside of the cancer arena, in a striking and concerning trend for 2009, innovation was conspicuous by its absence…”  Acknowledging this situation, there appears to be an increasing openness in the pharma industry for new approaches to drug discovery, sharing of data and collaboration with research groups and other parties, forming and joining health research consortia, and even for sharing of earlier-stage basic research information with competing pharma companies.  In other words, big pharma companies are to some extent joining into the overall collaborative game that will create PPPPM.

Diversity of participating parties. 

Diversity in the parties working to create PPPPM can be seen in size as well as type.  Included are international agencies, large and small pharma and biotech companies, health and research government agencies on the national, state, regional and community levels, world-scale computer, telecom and networking companies, software companies, data storage companies, supercomputer makers, data storage and chip companies,  medical schools and schools of public health, university , private and hospital research labs, computational chemistry groups, trade associations, healthcare associations, clinics, ambulatory care centers, physicians’ offices, long-term care facilities, insurance companies, HMOs, PPOs, genome scanning companies, data mining organizations, document management companies, regulatory compliance companies, small specialized companies of many additional kinds, big and small consulting companies and individual consultants and even writers like myself.

Inter-institutional collaboration. 

Collaborative networks and consortia that are precursors of PPPPM include many different kinds of participants.  I do not mean to suggest that there is a giant orgy of all of the kinds of organization I mentioned above working with all of the others.  However, many of the relevant emerging networks and consortia are rich in the kinds of organizations that are participating in them  An example is the PACeR consortium in New York State that combines hospitals, medical centers, healthcare networks, medical schools, pharma companies and healthcare associations, with a purpose being to significantly accelerate the clinical trials process.  I discussed another example of a multi-institutional health network last year in the blog entry The PROOF Centre of Excellence.  “The PROOF Centre is a cross-disciplinary engine of devoted partners including industry, academia, health care, government, patients and the public focused on reducing the enormous socioeconomic burdens of heart, lung and kidney failure and on improving health.” 

In silico. 

Biological R&D has traditionally taken place in-vitro (in the laboratory) and in-vivo (in living organisms).  In-silico narrowly refers to R&D that takes place in computers.  For example, a great deal about safety of a proposed new drug can be learned through applying computer models of toxicology and molecular biology to drug molecular structures, thus offering the possibility of significantly abbreviating Phase I clinical trials.  In-silico research can be used to correlate clinical outcomes information from numerous databases with analyzed gene and chromosomal information and gene expression data.  It can be used to “Examine mutation, copy number, expression and DNA methylation data from a wide variety of projects” and for a wide variety of other research purposes(ref).” 

More broadly, much of what will make PPPPM possible will take place in-silico including database creation, collaborative R&D, technical information-sharing among parties, distributed databases, online conferencing, etc.  The information storage and processing challenges of PPPM are mind-boggling.  Moore’s Law is still in operation after more than 50 years and should be good for at least another 10-15 years.  The law describes how computer power at any given price point doubles every two years.  Roughly the same is true for data storage capacity.  These two underlying factors are fundamental driving forces that are making PPPPM feasible.  The task is absolutely daunting and formidable, but the constant increase in silicon power that is being brought to bear on the task is equally formidable. 

Next generation sequencing

Next generation sequencing refers to a set of different high-throughput technologies for genome sequencing, recently extending to sequencing of transcriptomes, proteomes and epigenomes. Although the concept of next-gen sequencing technology has been around for some 5 years now, it continues to evolve through new generations  This video provides a nice background on sequencing technologies and applications.  The Illumina platforms provide a current standard for much current genome scanning.  CLC bio offers a turnkey system which sequences 32 full human genomes or 600 full human transcriptomes per week.

Petabyte storage requirements. 

The data volumes required to realize PPPM are staggering, unthinkable by traditional standards, today measured in petabytes.  A petabyte (derived from the SI prefix peta- ) is a unit of information equal to one quadrillion (short scale) bytes, or 1000 terabytes.”  To hold a petabyte you would need 250,000 4-gigabyte thumb drives.  The text in all the 33.32 million books in the library of congress is only a small fraction of a petabyte. Yet a major genome-scanning center can easily generate a petabyte of data every week or so.  A typical genome scan may generate 15 terabytes of raw data.  However, “in research facilities, raw sequence data is commonly kept for reinterpretation, and often includes redundant sets of data for the same genome (“fold coverage”). This increases the data storage and manipulation hardware needed for the already considerable output of a single sequencing run from the newest machines(ref).”   In a few years large data collections relevant to PPPPM will probably be measured in exabytes, where an exabyte is 1,000 petabytes.  The very large amounts of data storage required for genomics data has interested companies that specialize in providing superscale storage solutions.  For example, a news release a few days ago related “DataDirect Networks (DDN), the world’s largest privately-held information storage company, today announced the Stanford University Center for Genomics and Personalized Medicine selected DDN technology to provide massive scientific workflow scalability to its gene sequencing research.” Big storage companies like EMC Corporation and smaller ones like Teradata participated in the Bio-IT World Expo.

Processing the immense data streams required to realize PPPPM requires application of unprecedented computer power, particularly to crunch genomic and other omic data.  Popular approaches to getting the job done include  massively parallel processing, networked computing and cloud computing.

Massively parallel processing

The supercomputer manufacturers are ready to step up to the plate.   For example SGI had an exhibit at the trade show promoting the use of its massively parallel supercomputers involving very large numbers of processors. “Altix® UV scales to extraordinary levels-up to 256 sockets (2,048 cores, 4096 threads) with architectural support to 262,144 cores (32,768 sockets). Support for up to 16TB of global shared memory in a single system image, enables Altix UV to remain highly efficient at scale for applications ranging from in-memory databases, to a diverse set of data and compute-intensive HPC applications(ref).”

Networked computing

Networked computing, sometimes called distributed computing, involves splitting a major computing tasks over many machines in a network, possibly up to thousands of such machines.  Many distributed computing approaches have been applied to biologic data, a commercial example of which is Digipede.

Cloud computing

Although still fuzzily defined, cloud computing involves computing where either or both of data resources and processing software may lie out somewhere on “the cloud,” that is on Internet or on a private Internet network.  Only a browser or proprietary interface may be required on a user’s own computer.  As more and more data is being generated, it becomes less and less feasible to have it all on a user’s own computer or even on the computers in his/her’s own organization.  Also, it becomes more economical to have the processing software in a center where a great part of the data is located.  Advantages of cloud computing can be not having to invest in your own massive databases and elaborate software, improved all around economics, and facilitating collaboration.  The US Office of Management and Budget has mandated that federal agencies go to the cloud first whenever possible as an alternative to installing a new system of their own.  A great many biomedical databases and software applications are already available on the cloud and genomic data is increasingly being analyzed on the cloud.

From medical records to life-pattern records. 

Medical records have traditionally been gathered as a result of hospital stays or HMO participation and are largely mute with respect to what happens between hospital stays or visits.  There is very little information documenting the longer-term health effects of prolonged drug treatments or hospital procedures or medical radiation exposure.  A true individual health record would start with a whole-genome and epigenomic characterization at birth, periodic rescanning of the epigenome, and careful documentation of the consequence of every major medical or health intervention and important personal event.  There is increasing recognition of this necessity, but implementation remains largely in the future

Scalability

Multidimensional scalability becomes of increasing importance as PPPPM precursor systems expand in amounts of data, from gigabytes to terabytes to petabytes, in varieties of data, in the number of users and kinds of users.  For example, a research consortium might start out with a requirement for linking to the patient record systems in two or three hospitals, even if the patient record systems are not compatible in data organization or scope of content.  If the approach is scalable it should allow incorporation of more and more hospitals and additional varieties of patient record systems.  True scalability would allow incorporation of thousands of hospitals and clinics with millions of patients.  Considerations that affect scalability are discussed below and include data curation, data ontology, and semantic normalization of data elements.

Integrating data resources

There are hundreds if not thousands of specialized electronic genomic and gene association study databases out there.  A challenge for the PPPPM vision is identifying how to bring the information in all of these databases together within a single data architecture so comparative analyses can become possible across databases.  The challenge is outlined in the 2008 publication Genomic Data Resources: Challenges and Promises.   Approaches to this problem were described at the Conference & Expo in a number of presentations by researchers.  And several vendors featured software products and interface tools that can be used to tackle facets of the problem.  For example, “NextBio Enterprise is a secure web-based solution for integrating corporate and public data from next-gen sequencing and microarray technologies. Our unique “correlation engine” pre-computes billions of significant data connections and enables researchers to intelligently mine this data in real-time. With NextBio Enterprise, corporate experimental data can be easily integrated with public data and explored within relevant biological and clinical contexts(ref).” 

A few key concepts related to integrating data resources are data mining, data curation, data ontology, semantic normalization, metadata, desktop virtualization, and computational knowledge engines.

Data mining

Data mining is “is the process of extracting patterns from large data sets by combining methods from statistics and artificial intelligence with database management.[3]   Effective mining of biomedical data often requires an ontological approach and use of semantic data normalization.  See for example Ontology-assisted database integration to support natural language processing and biomedical data-mining.  “Successful biomedical data mining and information extraction require a complete picture of biological phenomena such as genes, biological processes and diseases as these exist on different levels of granularity. To realize this goal, several freely available heterogeneous databases as well as proprietary structured datasets have to be integrated into a single global customizable scheme.”

Data curation

There was much mention of data curation at the Conference. While the idea of curation seems to have been borrowed from the world of museums, it does seem to well-characterize what has to be done if we are going to see the PPPPM vision realized.Digital curation is the selection, preservation, maintenance, collection and archiving of digital assets[1][2] Digital curation is the process of establishing and developing long term repositories of digital assets for current and future reference[1] by researchers, scientists, and historians, and scholars generally(ref).” 

Data ontology

Ontology is recognized branch of philosophy and a wonderful abstract concept. I found it amazing to hear so much mention of it among both scientists and vendors in booths at a trade show.  Ontology (from the Greek ὄν, genitive ὄντος: “of that which is”, and -λογία, -logia: science, study, theory) is the philosophical study of the nature of being, existence or reality as such, as well as the basic categories of being and their relations. Traditionally listed as a part of the major branch of philosophy known as metaphysics, ontology deals with questions concerning what entities exist or can be said to exist, and how such entities can be grouped, related within a hierarchy, and subdivided according to similarities and differences(ref).”  The term is an excellent one:  what kinds of entities are represented by data elements in an epigenomic database, genomic disease association database or other component of an eventual vast PPPPM system?  How do we know those entities really exist?  What is their nature?  How do they relate? 

The idea of ontology-based data management for biomedical research databases goes back a number of years.  For background, you could have a look at the 2007 publication Ontology based data management systems for post-genomic clinical trials within a European Grid Infrastructure for Cancer Research and the 2010 publication The OBO Foundry: coordinated evolution of ontologies to support biomedical data integration.

Semantic normalization and metadata

Closely relate to the data ontology issue is that of semantic data normalization, that is, establishing equivalencies of meaning of data elements in different databases so meaningful analyses can be conducted across them.  Again closely associated is the concept of using metadata to establish such equivalencies, metadata being data about data.  From the previous citation: “Data management in post-genomic clinical trials is the process of collecting and validating clinical and genomic data with the goal to answer research questions and to preserve it for future scientific investigation. Comprehensive metadata describing the semantics of the data are needed to leverage it for further research like cross-trial analysis. Current clinical trial management systems mostly lack sufficient metadata and are not semantically interoperable. This paper outlines our approach to develop an application that allows trial chairmen to design their trial and especially the required data management system with comprehensive metadata according to their needs, integrating a clinical trial ontology into the design process.”

Desktop virtualization

Desktop virtualization is an approach used when effective analysis requires much more to be displayed on your computer “desktop” screen than can fit at any one time.  In its simplest form it is a way of rapidly shifting between views of information or data or data models.  Desktop virtualization is of increasing importance in health applications and is often combined with cloud computing.  And it is another component of the IT systems that are predecessors of PPPPM. For example, an announcement last week was Dell Launches Meditech Desktop Virtualization.  The mobile clinical computing system uses VMware technology to allow desktop healthcare applications to be accessed in the cloud and delivered as a managed service. — Dell has unveiled a mobile clinical computing (MCC) program that will enable healthcare organizations using the Meditech Health Care Information System (HCIS) to more easily and securely retrieve clinical information on their virtual desktops, as well as reduce desktop management, support, and deployment costs. — Announced Monday, the Meditech MCC program relies on a virtual desktop infrastructure (VDI) that is cost efficient, improves data management, and provides flexible deployment of virtual desktops in hospitals and their extended communities. — The program uses VMware vSphere, a cloud operating system, and VMware View, which allows desktop applications to be accessed in the cloud and delivered as a managed service. Additionally the solution includes Imprivata’s OneSign application and Forward Advantage’s API integration with the Meditech HCIS for using advanced authentication devices for e-signature actions. –In providing a technology solution for Meditech users, which include more than 2,300 hospitals, ambulatory care centers, physicians’ offices, and long-term care facilities, the MCC program is Dell’s latest attempt to explore new ways to offer technology to the healthcare.  sector.” Desktop virtualization is a particularly important issue for mobile applications where interfaces are displayed on small smartphone or tablet screens.

Movement from text searching to use of “computational knowledge engines”

Google and free text searching has done a wonderful job of bringing the world’s literature to every desktop and soon to every smartphone and other mobile device.  A new paradigm of knowledge retrieval (in contrast to information retrieval) may be emerging as exemplified by the WolframAlpha service now on the web.  WolframAlpha links a retrieval interface to the Woldfram Mathematica automated mathematical resources to provide answers to quantitative queries.  There is an attempt to decode the meaning of a query and arrive at an answer through analysis.  Right now WolframAlpha’s capabilities are severely limited.  It will, however, answer some questions like “How many base pairs in the human genome?” and “Number of deaths from malignant neoplasms?”  and “Average age of Parkinson’s Disease patients?” Try it!  As time goes on we can probably expect to see more and more sophisticated “knowledge engines” as applied to biomedical data. Some commercial and academic engines of this kind exist today.  

Linguamatics’ text mining software provides an example.  “Linguamatics I2E text mining software “provides that missing link – presenting the user with a text mining solution which enables very specific searches to be created on the fly (e.g. MAPK interacts with which targets?) and tabulated results to be returned very quickly. As well as generating data for analysis, this functionality enables I2E to be used in real time data-gathering tasks such as ontology creation which are becoming increasingly important in this sector.”

Everything I have discussed here is furthering the materialization of the expanded vision of PPPPM.  I believe that we are in the initial stages of creating a tectonic shift that profoundly enhances our health and longevity.  That shift probably won’t be called PPPPM, but whatever it is called it will propel us beyond the current stage where health maintenance is equated with taking drugs or medical treatment.  The shift will be made feasible by advances in information technology and come about through massive collaboration among multiple sectors of our society.

Note that I have no formal relationship with Cambridge Healthtech Institute, the organization that sponsors the Bio-IT World Conference and expo.  I have no relationship with any vendor or service-provider organization mentioned here and I receive no form of compensation for any such mention. 

Posted in Uncategorized | Leave a comment

Revisions to my dietary supplement firewall regimen

An important part of my treatise ANTI-AGING FIREWALLS – THE SCIENCE AND TECHNOLOGY OF LONGEVITY drafted in May 2008 has been the dietary supplement regimen. This post announces a new set of revisions to that regimen based on recent research results, mainly changes with respect to folic acid, PQQ, glucosamine, and phosphatidylcholine.
The treatise and the dietary supplement regimen
The basic organizing concept followed in writing the treatise was: 1. To identify and characterize the major existing theories of what causes aging, accepting that those theories are far from independent of each other. 2. For each theory of aging, assuming that theory is correct to identify what can practically be done to slow, halt, or even possibly reverse aging according to that theory. Specifically to identify lifestyle patterns and dietary supplements that could be taken to combat aging according to that theory. For example, given the oxidative damage theory of aging, lifestyle patterns would include avoiding many toxic substances like heavy metals and the suggested dietary supplements would include antioxidants. Thus I introduced for each theory of aging the idea of a lifestyle firewall and a dietary supplement firewall. 3. Finally, I combined the lifestyle firewalls for all the theories of aging to get a combined lifestyle regimen, and I combined the dietary supplement firewalls for the different theories of aging to get a combined dietary supplement regimen.
I have revised, updated and expanded the treatise every few weeks since writing it. But this basic organization still exists. While I started out with 14 basic theories of aging, in the course of time I added 6 more to the treatise and have discussed several additional ones in this blog. The suggestions in the dietary supplement regimen are based on own independent reviews of research. That is, each supplement is in the regimen only because I have read specific independent research publications, usually multiple ones, that have convinced me that the supplement exercises credible health-producing or anti-aging effects based both on both solid theoretical grounds and demonstrable experimental results. Because the theories of aging are not independent of each other many of the suggested dietary supplements are protective of health and against aging according to multiple theories. My firewall approach to longevity is a simple and practical one. When I drafted the first version of this document in 2008, I saw this approach as appropriate for older people given the current state of knowledge and our relative ignorance compared to what will be known in coming decades. And I still see it that way. However, my views have been far from static. As I have learned more about the sciences of longevity and the overall state of knowledge has expanded, I have continued to revise the firewalls regimens. For example, by November of 2010 there was more published knowledge about telomerase biology and telomerase and the depth of my personal knowledge about this field had increased very significantly. Based on this knowledge I decided to discontinue use of cycloastragenol or any other specialized commercial “telomerase activator.” See the March 2011 blog entry The epigenetic regulation of telomeres.
I announce and explain the new changes here. My regular readers have possibly seen a few of these coming.
Folic acid
I am keeping this supplement in the regimen at a level of 900mcg a day but with a caution that a suggested daily dose of 1,200 mcg not be exceeded. This could happen, for example, by taking a folic acid supplement and two B-complex capsules daily, each of which contain 900mcg of folic acid. The research that has led me to this shift is detailed in the February 2011 blog entry The many faces of folic acid.
PQQ
On reflection after generating the April 2011 blog entry PQQ – activator of PGC-1alpha, SIRT3 and mitochondrial biogenesis, I have decided to commence supplementation with PQQ at a level of 10mg per day. The health and longevity-benefits research case for doing this are described in the blog entry.
Glucosamine
I am keeping this supplement in the regimen at level of 1,500 mg a day but with caution suggesting that daily dosage not exceed that level.
This decision is a consequence of a few quite-recent studies. The first was published in October 2010 which points out a couple if important negative consequences of consumption of glucosamine: decreased SIRT1 levels and apoptosis of pancreatic cells. The publication is Hexosamines stimulate apoptosis by altering Sirt1 action and levels in rodent pancreatic beta-cells. “The activity and levels of Sirt1, which promotes cell survival in several models, are linked to glucose concentrations and cellular energy metabolism. The present study aimed at determining whether impaired Sirt1 activity is involved in the induction of apoptosis by the nutrient-sensing hexosamine biosynthesis pathway (HBP). Pancreatic Nit-1, Rinf-m5F and Min6 beta-cells were acutely treated at different doses and times with glucosamine, which directly enters and stimulates the HBP. Sirt1 levels were genetically modulated by retroviral infection. Expression levels, cellular localization, and activity of apoptosis-related markers were determined by qPCR, immunoblotting and co-immunoprecipitation. Glucosamine dose- and time-dependently induced cell apoptosis in all cell lines studied. HBP stimulation time-dependently modified Sirt1 protein levels, notably in the cytoplasm. This was concomitant with increased E2F1 binding to the c-myc promoter. In both NIT-1 and min6 beta-cells, genetic knockdown of Sirt1 expression resulted in higher susceptibility to HBP-stimulated apoptosis, whereas overexpression of Sirt1 had the opposite impact. These findings indicate that reduction of Sirt1 levels by hexosamines contributes to beta-cell apoptosis.”
A discussion of the implications of this research can be found in the October 2010 Science Daily article Too Much Glucosamine Can Cause the Death of Pancreatic Cells, Increase Diabetes Risk, Researchers Find. “In vitro tests conducted by Professor Frédéric Picard and his team revealed that glucosamine exposure causes a significant increase in mortality in insulin-producing pancreatic cells, a phenomenon tied to the development of diabetes. Cell death rate increases with glucosamine dose and exposure time. “In our experiments, we used doses five to ten times higher than that recommended by most manufacturers, or 1,500 mg/day,” stressed Professor Picard. “Previous studies showed that a significant proportion of glucosamine users up the dose hoping to increase the effects,” he explained. — Picard and his team have shown that glucosamine triggers a mechanism intended to lower very high blood sugar levels. However, this reaction negatively affects SIRT1, a protein critical to cell survival. A high concentration of glucosamine diminishes the level of SIRT1, leading to cell death in the tissues where this protein is abundant, such as the pancreas. — Individuals who use large amounts of glucosamine, those who consume it for long periods, and those with little SIRT1 in their cells are therefore believed to be at greater risk of developing diabetes. In a number of mammal species, SIRT1 level diminishes with age. This phenomenon has not been shown in humans but if it were the case, the elderly — who constitute the target market for glucosamine — would be even more vulnerable. — “The key point of our work is that glucosamine can have effects that are far from harmless and should be used with great caution,” concluded Professor Picard.””
A December 2010 publication O-GlcNAc modification, insulin signaling and diabetic complications speaks to a form of glycation damage which could possibly be exacerbated by intake of dietary glucosamine. “O-GlcNAc glycosylation (O-GlcNAcylation) corresponds to the addition of N-acetylglucosamine on serine and threonine residues of cytosolic and nuclear proteins. O-GlcNAcylation is a dynamic post-translational modification, analogous to phosphorylation, that regulates the stability, the activity or the subcellular localisation of target proteins. This reversible modification depends on the availability of glucose and therefore constitutes a powerful mechanism by which cellular activities are regulated according to the nutritional environment of the cell. O-GlcNAcylation has been implicated in important human pathologies including Alzheimer disease and type-2 diabetes. Only two enzymes, OGT and O-GlcNAcase, control the O-GlcNAc level on proteins. Therefore, O-GlcNAcylations cannot organize in signaling cascades as observed for phosphorylations. O-GlcNAcylations should rather be considered as a “rheostat” that controls the intensity of the signals traveling through different pathways according to the nutritional status of the cell. Thus, OGT attenuates insulin signal by O-GlcNAcylation of proteins involved in proximal and distal steps in the PI-3 kinase signaling pathway. This negative feedback may be exacerbated when cells are chronically exposed to elevated glucose concentrations and could thereby contribute to alterations in insulin signaling observed in diabetic patients. O-GlcNAcylation also appears to contribute to the deleterious effects of hyperglycaemia on excessive glucose production by the liver and deterioration of β-cell pancreatic function, resulting in worsening of hyperglycaemia (glucotoxicity). Moreover, O-GlcNAcylations directly participate in several diabetic complications. O-GlcNAcylation of eNOS in endothelial cells have been involved in micro- and macrovascular complications. In addition, O-GlcNAcylations activate the expression of profibrotic and antifibrinolytic factors, contributing to vascular and renal dysfunctions.”
An April 2011 publication casts another dark shadow on the potential impact of glucosamine supplementation: Oral glucosamine increases expression of transforming growth factor β1 (TGF β 1) and connective tissue growth factor (CTGF) mRNA in rat cartilage and kidney: Implications for human efficacy and toxicity. “Lean Zucker rats were dosed orally for 6 weeks with glucosamine hydrochloride at doses (0 – 600 mg/kg/day) that produced peak serum concentrations of <1 – 35 μM, spanning the human exposure range. Relative expression of both TGFβ1 and CTGF mRNA were significantly increased up to 2.3-fold in liver, kidney and articular cartilage when evaluated 4 hours after final dose. Apparent threshold serum glucosamine (C(max)) concentration required to increase TGFβ1 expression in cartilage was 10 – 20 μM. These increases were associated with significant increases in UDP-N-acetylglucosamine concentrations suggesting increased hexosamine flux. Both TGFβ1 and CTGF are mediators of chondrocyte proliferation and cartilage repair. Study demonstrates that oral glucosamine doses that produce clinically relevant serum glucosamine concentrations can induce tissue TGFβ1 and CTGF expression in vivo and provides a mechanistic rationale for reported beneficial effects of glucosamine therapy. Induction of renal TGFβ1 and CTGF mRNA suggests that potential sclerotic side-effects may occur following consumption of potent glucosamine preparations.”

Together, these studies suggest that negative health and longevity effects could well result from the prolonged taking of large doses of glucosamine.

Efficacy of glucosamine for averting or treating osteoarthritis is another matter with some studies supporting its capability to address joint diseases and other studies suggesting that the existence of such a capability is questionable. The last study cited above suggests it may be efficacious.   Also in that category is the work reported in the March 2011 publication Effects of glucosamine derivatives and uronic acids on the production of glycosaminoglycans by human synovial cells and chondrocytes. “Glucosamine (GlcN) has been widely used to treat osteoarthritis (OA) in humans. However, its chondroprotective action on the joint is poorly understood. In this study, to elucidate the chondroprotective action of GlcN, we examined the effects of GlcN-derivatives (GlcN and N-acetyl-D-glucosamine) and uronic acids (D-glucuronic acid and D-galacturonic acid) (0.1-1 mM) on the production of glycosaminoglycans (GAG), such as hyaluronic acid (HA), keratan sulfate and sulfated GAG by human synovial cells and chondrocytes. — Together these observations indicate that GlcN may exhibit chondroprotective action on joint diseases such as OA by modulating the expression of HA-synthesizing enzymes and inducing the production of HA (a major component of GAG contained in synovial fluid) especially by synovial cells.”

 
Phosphatidylcholine
I am discontinuing inclusion of phosphatidylcholine (lecithin) in the dietary supplement regimen.
Phosphatidylcholine (PC) are a class of phospholipids that incorporate choline as a headgroup. They are a major component of biological membranes and can be easily obtained from a variety of readily available sources such as egg yolk or soy beans from which they are mechanically extracted or chemically extracted using hexane. They are also a member of the lecithin group of yellow-brownish fatty substances occurring in animal and plant tissues(ref).”
The proximate reason for eliminating phosphatidylcholine from the regimen are laid out in the April 2011 publication Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. “Metabolomics studies hold promise for the discovery of pathways linked to disease processes. Cardiovascular disease (CVD) represents the leading cause of death and morbidity worldwide. Here we used a metabolomics approach to generate unbiased small-molecule metabolic profiles in plasma that predict risk for CVD. Three metabolites of the dietary lipid phosphatidylcholine—choline, trimethylamine N-oxide (TMAO) and betaine—were identified and then shown to predict risk for CVD in an independent large clinical cohort. Dietary supplementation of mice with choline, TMAO or betaine promoted upregulation of multiple macrophage scavenger receptors linked to atherosclerosis, and supplementation with choline or TMAO promoted atherosclerosis. Studies using germ-free mice confirmed a critical role for dietary choline and gut flora in TMAO production, augmented macrophage cholesterol accumulation and foam cell formation. Suppression of intestinal microflora in atherosclerosis-prone mice inhibited dietary-choline-enhanced atherosclerosis. Genetic variations controlling expression of flavin monooxygenases, an enzymatic source of TMAO, segregated with atherosclerosis in hyperlipidaemic mice. Discovery of a relationship between gut-flora-dependent metabolism of dietary phosphatidylcholine and CVD pathogenesis provides opportunities for the development of new diagnostic tests and therapeutic approaches for atherosclerotic heart disease.”
pdc-1.jpg

A Eurekalert writeup of this research had to say: “A new pathway has been discovered that links a common dietary lipid and intestinal microflora with an increased risk of heart disease, according to a Cleveland Clinic study published in the latest issue of Nature. — The study shows that people who eat a diet containing a common nutrient found in animal products (such as eggs, liver and other meats, cheese and other diary products, fish, shellfish) are not predisposed to cardiovascular disease solely on their genetic make-up, but rather, how the micro-organisms that live in our digestive tracts metabolize a specific lipid — phosphatidyl choline (also called lecithin). Lecithin and its metabolite, choline, are also found in many commercial baked goods, dietary supplements, and even children’s vitamins. — The study examined clinical data from 1,875 patients who were referred for cardiac evaluation, as well as plasma samples from mice. When fed to mice, lecithin and choline were converted to a heart disease-forming product by the intestinal microbes, which promoted fatty plaque deposits to form within arteries (atherosclerosis); in humans, higher blood levels of choline and the heart disease forming microorganism products are strongly associated with increased cardiovascular disease risk. — “When two people both eat a similar diet but one gets heart disease and the other doesn’t, we currently think the cardiac disease develops because of their genetic differences; but our studies show that is only a part of the equation,” said Stanley Hazen, M.D., Ph.D., Staff in Lerner Research Institute’s Department of Cell Biology and the Heart and Vascular Institute’s Department of Cardiovascular Medicine and Section Head of Preventive Cardiology & Rehabilitation at Cleveland Clinic, and senior author of the study. “Actually, differences in gut flora metabolism of the diet from one person to another appear to have a big effect on whether one develops heart disease. Gut flora is a filter for our largest environmental exposure – what we eat.” — Dr. Hazen added, “Another remarkable finding is that choline – a natural semi-essential vitamin – when taken in excess, promoted atherosclerotic heart disease. Over the past few years we have seen a huge increase in the addition of choline into multi-vitamins – even in those marketed to our children – yet it is this same substance that our study shows the gut flora can convert into something that has a direct, negative impact on heart disease risk by forming an atherosclerosis-causing by-product.” — In studies of more than 2,000 subjects altogether, blood levels of three metabolites of the dietary lipid lecithin were shown to strongly predict risk for cardiovascular disease: choline (a B-complex vitamin), trimethylamine N-oxide (TMAO, a product that requires gut flora to be produced and is derived from the choline group of the lipid) and betaine (a metabolite of choline). — “The studies identify TMAO as a blood test that can be used in subjects to see who is especially at risk for cardiac disease, and in need of more strict dietary intervention to lower their cardiac risk,” Dr. Hazen said. — Healthy amounts of choline, betaine and TMAO are found in many fruits, vegetables and fish. These three metabolites are commonly marketed as direct-to-consumer supplements, supposedly offering increased brain health, weight loss and/or muscle growth. — These compounds also are commonly used as feed additives for cattle, poultry or fish because they may make muscle grow faster; whether muscle from such livestock have higher levels of these compounds remains unknown.”
Even prior to encountering this publication I had been uncomfortable with one aspect of phosphatidylcholine: phosphatidylcholine stimulates the expression of NF-kappaB(ref)(ref)(ref). My treatise discusses how aberrant NF-kappaB signaling appears to characterize many inflammatory disease processes. They include promotion of angiogenesis, proliferation, metastasis and invasiveness in cancer tumors, autoimmune diseases, neurodegenerative diseases and contributing to the activation of human immunodeficiency virus (HIV) leading to AIDS. — There appears to be increasing evidence that inhibition of expression of NF-kB could be a key approach for fighting cancers, controlling inflammatory diseases, AIDS, neurodegenerative conditions like Parkinson’s Disease and a number of other significant age-related maladies.” So promotion of expression of NF-kappaB is the opposite of what is wanted. In this respect, phosphatidylcholine has behaved in an opposite manner to thirty-nine pluripotent substances in my dietary regimen which suppress the expression of NF-kappaB.
This information and the other information offered in this blog or in the associated treatise is not intended for diagnosis or treatment of any medical condition and should not be construed as medical advice. The information is not a substitute for a licensed physician’s medical advice. If any opinions or suggestions herein conflict with that of a treating licensed physician, defer to the opinion of the physician. This information is intended for people in good health. The dietary supplement regimen described herein is that used by the author and may not be appropriate for other people. It is the user’s responsibility to know his or her health and medical history and ensure that supplements he or she takes do not create an adverse reaction.
The author does not have nor has ever had any business relationship with any company that packages or markets any dietary supplement.

Posted in Uncategorized | 20 Comments

PQQ – activator of PGC-1alpha, SIRT3 and mitochondrial biogenesis

By Vince Giuliano 

Minor update July 12, 2013

Pyrroloquinoline quinone (PQQ), a redox cofactor available as a dietary supplement, appears to have at least three central biological effects with powerful downstream health and longevity consequences: it stimulates the generation of PGC1-alpha, results in expression of SIRT3, and induces mitochondrial biogenesis.  After brief recapitulation of background, I review some of the key research literature involved.  Of possible practical significance, supplementation with PQQ could possibly offer the benefits of exercise in a pill.

 Background on PGC-1alpha

In a June 2010 blog entry AMPK and longevity, I discussed how exercise activates the AMPK pathway and the role of PGC-1alpha (peroxisome-proliferator-activated receptor gamma co-activator-1alpha) as a co-transcriptional regulation factor that induces mitochondrial biogenesis by activating transcription factors.

The August 2010 blog entry PGC-1alpha and exercise provides a further and more general introduction to PGC1alpha.  I said “You can probably expect to hear a lot about PGC-1alpha as time goes on because this remarkable substance is turning out to have a lot to do with health and longevity. It appears to be the mediator of the health benefits produced by exercise. This blog post is about PGC-1alpha, about its relationship to exercise, and about efforts to stimulate it with various substances, in essence seeing if it is possible to provide “exercise in a pill.” 

In that blog entry I described how exercise generates PGC1alpha. Expression of PGC-1alpha can also come about as a consequence of cellular stress such as induced by cold.  I also discussed a number of physiological properties of PGC-1alpha affecting health and longevity including:

·        PGC-1alpha regulates energy metabolism involving both white and brown fat.  PPARgamma coactivator-1alpha (PGC-1alpha), in cooperation with several transcription factors, has emerged as a key regulator of several aspects of mammalian energy metabolism including mitochondrial biogenesis, adaptive thermogenesis in brown adipose tissue, glucose uptake, fiber type-switching in skeletal muscle, gluconeogenesis in liver and insulin secretion from pancreas. Recent studies have shown a reduced expression of PGC-1alpha in skeletal muscle of diabetic and prediabetic humans. Moreover, expression of PGC-1alpha in white fat cells activates a broad program of adaptive thermogenesis characteristic of brown fat cells(ref).”

·         PGC-1alpha turns on the biogenesis of mitochondria primarily in brown fat, working through NRF1, NRF2 and ERRalpha. It promotes fatty acid oxidation working through the PPARs and RXRs, NRF1 and NRF2, combats ROS and promotes glucose utilization, promotes oxidative phosphorylation working via NRF2 and ERRalpha, promotes angiogenesis working through ERRalpha, and contributes to fiber-type switching.”

·        PGC-1alpha stimulates mitochondrial biogenesis and promotes the remodeling of muscle tissue to a fiber-type composition that is metabolically more oxidative and less glycolytic in nature, and it participates in the regulation of both carbohydrate and lipid metabolism(ref).”

·        Exercise-induced expression of PGC-1alpha appears to enhance insulin sensitivity.  Thus, it is likely that maintenance of upregulated levels of PGC-1alpha is protective against diabetes.

·        Muscle PGC-1alpha protects against oxidative damage in aging muscle and PGC1alpha prevents age-related loss of endurance running capacity.

·        “It is highly likely that PGC-1alpha is intimately involved in disorders such as obesity, diabetes, and cardiomyopathy. In particular, its regulatory function in lipid metabolism makes it an inviting target for pharmacological intervention in the treatment of obesity and Type 2 diabetes(ref).”

Background on sirt3 mitochondrial biogenesis

Besides the above references to mitochondrial biogenesis, the blog entry SIRT3 research – tying together knowledge of aging points out:

·        Systematic exercise up-regulates PGC-1alpha and increases SIRT3 expression as well as associated CREB phosphorylation. “CREB (cAMP response element-binding) is a cellular transcription factor. It binds to certain DNA sequences called cAMP response elements (CRE), thereby increasing or decreasing the transcription of the downstream genes[1] (ref).”

·        Cellular stress causes SIRT3 to translocate from the nucleus to the mitochondria and to be highly expressed in brown adipose tissue.  SIRT3 is a mitochondrial sirtuin protein that serves to deacetylate acetyllysine-modified proteins in mitochondria.  

·        “(PGC-1alpha) plays important roles in adaptive thermogenesis, gluconeogenesis, mitochondrial biogenesis and respiration. PGC-1alpha induces several key reactive oxygen species (ROS)-detoxifying enzymes, –Here we show that PGC-1alpha strongly stimulated mouse Sirt3 gene expression in muscle cells and hepatocytes. – Furthermore, Sirt3 was essential for PGC-1alpha-dependent induction of ROS-detoxifying enzymes and several components of the respiratory chain, including glutathione peroxidase-1, superoxide dismutase 2, ATP synthase 5c, and cytochrome c. — Our results indicate that Sirt3 functions as a downstream target gene of PGC-1alpha and mediates the PGC-1alpha effects on cellular ROS production and mitochondrial biogenesis. Thus, SIRT3 integrates cellular energy metabolism and ROS generation(ref).”

 Background on PQQ

PQQ is a quinone, a bacterial redox co-factor and antioxidant.  It is a natural dietary substance found in common foods, good sources being parsley, green tea, tofu, tomatoes green peppers, kiwi fruit and papaya(ref).The roots of the original discovery of PQQ goes back to 1964(ref) and the substance was first isolated in 1979(ref).  However, the biological significance of the substance has been discovered only through a chain of slowly-developing research.  Much of the original research on PQQ was concerned with its roles in bacteria.  PQQ was not known to be essential to human metabolism until 2003 and research on its central roles in stimulating PGC-1alpha and mitochondrial biogenesis was published until 2010.  The Wikipedia article on PQQ provides an excellent overview with an ample list of research citations. 

The basic relevance of PQQ in terms of the above discussion, put simply, is that it is an upstream activator of CREB which is an activator of PGC-1alpha which in turn activates SIRT3.  Therefore, PQQ is potentially an initiator of the chain of health and longevity benefits that devolve from expression of PGC-1alpha and SIRT3.  The substance could be the long sought-after “exercise in pill form.”

There appear to be some 200 publications on PQQ including a few books.  I will skip over the historical publications and start with a key 2010 publication and work selectively forwards from there.  That 2010 publication is Pyrroloquinoline quinone stimulates mitochondrial biogenesis through cAMP response element-binding protein phosphorylation and increased PGC-1alpha expression.  Bioactive compounds reported to stimulate mitochondrial biogenesis are linked to many health benefits such increased longevity, improved energy utilization, and protection from reactive oxygen species. Previously studies have shown that mice and rats fed diets lacking in pyrroloquinoline quinone (PQQ) have reduced mitochondrial content. Therefore, we hypothesized that PQQ can induce mitochondrial biogenesis in mouse hepatocytes. Exposure of mouse Hepa1–6 cells to 10–30 μm PQQ for 24–48 h resulted in increased citrate synthase and cytochrome c oxidase activity, Mitotracker staining, mitochondrial DNA content, and cellular oxygen respiration. The induction of this process occurred through the activation of cAMP response element-binding protein (CREB) and peroxisome proliferator-activated receptor-gamma coactivator-alpha (PGC-1alpha), a pathway known to regulate mitochondrial biogenesis. PQQ exposure stimulated phosphorylation of CREB at serine 133, alpha mRNA and protein expression. PQQ did not stimulate mitochondrial biogenesis after small interfering RNA-mediated reduction in either PGC-1alpha or CREB expression. Consistent with activation of the PGC-1alpha pathway, PQQ increased nuclear respiratory factor activation (NRF-1 and NRF-2) and Tfam, TFB1M, and TFB2M mRNA expression. Moreover, PQQ protected cells from mitochondrial inhibition by rotenone, 3-nitropropionic acid, antimycin A, and sodium azide. The ability of PQQ to stimulate mitochondrial biogenesis accounts in part for action of this compound and suggests that PQQ may be beneficial in diseases associated with mitochondrial dysfunction.”

It is established that cAMP response element-binding protein (CREB) is an important regulator of PGC-1alpha that facilitates PGC-1alpha activation (ref).

NRF-2 is introduced in the blog entry Nrf2 and cancer chemoprevention by phytochemicals and I have mentioned its beneficial effects in a number of other blog postings.  “Nuclear factor-erythroid-2-related factor 2 (Nrf2) plays a crucial role in the coordinated induction of those genes encoding many stress-responsive and cytoptotective enzymes and related proteins. These include NAD(P)H:quinone oxidoreductase-1, heme oxygenase-1, glutamate cysteine ligase, glutathione S-transferase, glutathione peroxidase, thioredoxin, etc.(ref).” (Several additional blog entries written after this one further characterize the health benefits of Nrf2.)

 The function of PQQ in mammalian physiology remains controversial. PQQ has been proposed as a vitamin (19), but it has not been demonstrated that PQQ serves as an enzyme cofactor in mammalian tissues (20, 21). Upon appreciation that mitochondrial content can be influenced by PQQ nutritional status and that reported beneficial effects of PQQ may be directly related to mitochondrial function, we hypothesized that PQQ may induce mitochondrial biogenesis through a mitochondrial-related cell signaling mechanism. Given that many mitochondrial-related events are regulated by PGC-1alpha and nuclear respiratory factors (15), we hypothesized that PQQ may interact with a PGC-1alpha-related pathway. We used the mouse Hepa1–6 hepatocyte cell line as a model to investigate these hypotheses. We also explored whether PQQ may protect against the toxic effects of mitochondrial electron transport chain inhibition(ref).”

The aforementioned article goes on to describe experimental procedures that establish:

·        PQQ induces mitochondrial biogenesis.

·        PQQ induces nuclear respiratory factor (NRF) activation, both NRF-1 and NRF-2.  

·        PQQ induces PGC-1alpha promoter activation and increases PGC-1alpha mRNA and protein expression.  Further, PGC-1alpha binding can directly regulate NRF-1 and NRF-2 activity.

·        PGC-1alpha is required for the induction of mitochondrial biogenesis by PQQ.

·        PQQ stimulates the phosphorylation of CREB at serine 133 and CREB is required for PQQ-induced mitochondrial biogenesis.

·        PQQ-mediated mitochondrial biogenesis is not due to auto-oxidation or IPQ addition.  That is, “PQQ-induced mitochondrial biogenesis and nuclear respiratory factor activation is not caused by generation of either hydrogen peroxide, superoxide, or IPQ in the media by PQQ(ref).” 

·        PQQ improves cell viability and preserves mitochondrial function due to mitochondrial inhibitors.

·        The exact role of CREB in  PQQ stimulation of  PGC-1alpha is not clear. “CREB has been shown to regulate PGC-1alpha, and data shown here suggest that PQQ acts through CREB to regulate PGC-1alpha, but regulation of the PGC-1alpha and PGC-1-related co-activator by CREB has not yet been demonstrated. Also, it is recognized that the PGC-1 family of co-activators likely act and respond to differing physiological stimuli and processes (ref).”

PQQ and neuroprotectivity

 The 2011 publication The neuroprotective action of pyrroloquinoline quinone against glutamate-induced apoptosis in hippocampal neurons is mediated through the activation of PI3K/Akt pathwayreports “In this study, we investigated the effects of PQQ on glutamate-induced cell death in primary cultured hippocampal neurons and the possible underlying mechanisms. We found that glutamate-induced apoptosis in cultured hippocampal neurons was significantly attenuated by the ensuing PQQ treatment, which also inhibited the glutamate-induced increase in Ca2+ influx, caspase-3 activity, and ROS production, and reversed the glutamate-induced decrease in Bcl-2/Bax ratio. The examination of signaling pathways revealed that PQQ treatment activated the phosphorylation of Akt and suppressed the glutamate-induced phosphorylation of c-Jun N-terminal protein kinase (JNK). — Taken together, our results indicated that PQQ could protect primary cultured hippocampal neurons against glutamate-induced cell damage by scavenging ROS, reducing Ca2+ influx, and caspase-3 activity, and suggested that PQQ-activated PI3K/Akt signaling might be responsible for its neuroprotective action through modulation of glutamate-induced imbalance between Bcl-2 and Bax.”

PQQ and uranium toxicity

The 2011 publication Uranium exerts acute toxicity by binding to pyrroloquinoline quinone cofactor provides at least one explanation of why uranium is so toxic – it binds to PQQ and prevents its biological actions.  “Uranium as an environmental contaminant has been shown to be toxic to eukaryotes and prokaryotes; however, no specific mechanisms of uranium toxicity have been proposed so far. Here a combination of in vivo, in vitro, and in silico studies are presented describing direct inhibition of pyrroloquinoline quinone (PQQ)-dependent growth and metabolism by uranyl cations. Electrospray-ionization mass spectroscopy, UV-vis optical spectroscopy, competitive Ca(2+)/uranyl binding studies, relevant crystal structures, and molecular modeling unequivocally indicate the preferred binding of uranyl simultaneously to the carboxyl oxygen, pyridine nitrogen, and quinone oxygen of the PQQ molecule. The observed toxicity patterns are consistent with the biotic ligand model of acute metal toxicity. In addition to the environmental implications, this work represents the first proposed molecular mechanism of uranium toxicity in bacteria, and has relevance for uranium toxicity in many living systems.”

Other health benefits of PPQ

Here is a sample of recent publications outlining other health benefits of PPQ, particularly as related to cancers and Alzheimer’s disease:

·        Pyrroloquinoline quinone inhibits the fibrillation of amyloid proteins. (2010)

·        Role of glutathione in augmenting the anticancer activity of pyrroloquinoline quinone (PQQ)  (2010)

·        Protective effect of pyrroloquinoline quinone against Abeta-induced neurotoxicity in human neuroblastoma SH-SY5Y cells  (2009)

·        Identification of transcriptional networks responding to pyrroloquinoline quinone dietary supplementation and their influence on thioredoxin expression, and the JAK/STAT and MAPK pathways  (2010)

·        The inhibitory effect of pyrroloquinoline quinone on the amyloid formation and cytotoxicity of truncated alpha-synuclein (2010)

In addition, there have been many recent studies concerned with the chemistry and biochemistry of PQQ and its behavior in a number of bacteria.

The case for PQQ as a dietary supplement

 The first-mentioned publication goes on in its discussion to make what seems to be a possible case for PQQ supplementation: “The induction of mitochondrial biogenesis by PQQ has a number of health implications. PGC-1α elevation, particularly in muscle and adipose tissue, may also be helpful in that PGC-1alpha expression is decreased in obesity (56, 57). CREB null and PGC-1alpha null mice have hepatic steatosis and impaired gluconeogenesis and β-oxidation (38, 58, 59). PQQ-deficient mice have elevated serum triglycerides, which is reversed upon PQQ repletion (1). In addition, mice with deletion of all CREB isoforms have reduced commissural structure formation and impaired fetal T cell development (60), and other mouse models of CREB-targeted deletion show impaired memory and neurodegeneration (61, 62). Likewise, dietary PQQ deprivation results in immune dysfunction (63). PQQ is also neuroprotective when administered by intraperitoneal injection (64, 65) or diet supplementation (66).”

PQQ is not the only dietary supplement that can activate PGC-1alpha and therefore unleash its chain of beneficial results including enhanced SIRT3 expression and mitochondrial biogenesis.  Resveratrol and and other substances in my anti-aging firewalls dietary supplement regimen, can also do this.  And the drug rapamycin can also enhance PGC-1alpha expression(ref).

The basic arguments for PQQ supplementation relate to solubility and bioavailability.  Going on to quote additionally from the 2010 publication: “Although other phytochemicals are associated with the activation of cell signaling pathways important to mitochondrial function, PQQ has properties that set it apart from other compounds. As an example, resveratrol and genisten have been demonstrated to affect cell-signaling pathways, including those important for mitochondrial biogenesis. Resveratrol can induce deacetylation of PGC-1alpha (ref) and AMP-activated protein kinase activation (ref), which are potential mechanisms for PGC-1α activation. Both resveratrol and genistein are relatively insoluble in water, and increasing its water solubility does not increase resveratrol absorption (ref), although genistein bioavailability can be increased by complexing genistein with cyclodextrins (ref). In contrast, PQQ is relatively water-soluble (>1 g of PQQ/liter of water) and is easily absorbed at low dietary concentrations intakes (ref). Although genistein can induce PGC-1alpha protein expression and mitochondrial biogenesis (3), genistein may also have phytoestrogenic properties because of its ability to activate the estrogen receptor (ref).”

Going on: “The observed effects of PQQ are also observed at concentrations lower than those for resveratrol and genistein, particularly in vivo. In cell cultures in vitro, PQQ causes changes in mitochondriogenesis and function at concentrations similar to those reported recently for small molecule activators of SIRT1 (ref), which are being explored for their therapeutic potential (ref). These observations suggest that further study related to PQQ is warranted. One important note is that PQQ can increase PGC-11alpha mRNA transcription, which is different from the post-translation regulation of PGC-1alpha by resveratrol and raises the likelihood that a combination of various compounds, such as are often present in fruits and vegetables, can stimulate mitochondrial biogenesis through different modes of action. Because mitochondria function as the principal energy source of the cell, compromised function of this key organelle is linked to numerous diseases and metabolic disorders (ref)(ref). In this regard, PQQ would appear to have therapeutic potential similar to resveratrol, genistein, hydroxytyrosol, quercetin, or other compounds that can induce mitochondrial biogenesis(ref).”

Does PGG supplementation make sense?

My impression is that the PQQ supplementation phenomenon is new and just taking off. A few of the supplement companies have picked up on the 2010 findings outlined above and are starting to offer PQQ supplements, Life Extension Foundation having been a leader in this regard.  Most supplement vendors offer PQQ only in proprietary combinations with other substances.   I have found this discussion of PPG supplementation dosage on a site providing practical Pyrroloquinoline Quinone & Methoxatin information. My impression is that the information on that site is responsible and informative although not completely documented.  For example, that site contains an  interesting table for the PQQ  content that researchers have found in food.  That discussion points out that there are many imponderable factors that could affect what is the optimum amount for a healthy adult to take as a daily PQQ supplement. “This question has not been fully resolved and is complicated by the inability to measure all forms of PQQ (pyrroloquinoline quinone | methoxatin). However, it is possible to make several good inferences from current animal studies and data–.”  The discussion argues that, all things considered, 10-20mg of PQQ a day is probably a reasonable dose.  The supplement-makers apparently concur and the PQQ supplement products I have seen range from having 5 to 20 mg of PQQ per pill. 

In considering PQQ supplementation, I hasten to point out that a number of other dietary supplements offer positive benefits for mitochondrial health including r-alpha-lipoic acid, acetyl-l-carnitine, resveratrol, quercetin and co-enzyme Q10.  For a discussion of the supplements I have been  taking related to mitochondrial health, you can have a look at the Mitochondrial Damage Firewall in my treatise.

Some supplement marketers are representing PQQ as being a new vitamin, although whether it satisfies the criteria for being a vitamin has been disputed(ref).

Given that I exercise at least 45 minutes a day, eat a reasonable diet, and take all the supplements in my anti-aging firewalls dietary supplement regimen, do I think additional enhancement of PGC-1alpha and all of the associated health benefits would come about for me if I added PQQ as a supplement?  Or would adding PQQ to my dietary regimen be simply redundant?  Or could I increase the effectiveness or reduce the cost of the regimen by substituting PQQ for other supplements?  I simply did not know when I first drafted this blog in April 2011.  Because of the importance of SIRT3 expression, however, in 2012 I started regular supplementation with PQQ and have continued that pattern since (July 2013).

Since drafting the above over two years ago,  another 50 research publications relating to PQQ have appeared in pubmed.org, further elucidating its mechanisms of action and health benefits. In the interim I have developed an increased appreciation of the importance of SIRT3 expression for controlling mitochondrial ROS and general health maintenance during aging.  At some point I will generate a new blog entry covering the new research and updating this one.

Posted in Uncategorized | 10 Comments

Personal protection against nuclear radioactive substances

The situation at the Fukushima nuclear plant in Japan is giving great urgency to the issue of personal protection against ionizing radiation.  This blog entry is about how a degree of personal protection against radiation damage can be achieved by taking antioxidants.  I do not think it is an exaggeration to say that what I will be discussing could in the long term save the lives of millions of people. 

The Fukushima nuclear disaster

As of this writing the situation at the Fukushima Daiichi reactors remains uncontrolled.   It is a slowly-unfolding disaster, appearing to get worse and worse.   Smoke or metallic vapor from overheated or melted fuel rods contain large quantities of radioactive fission byproducts.   And very little can be done to stop fuel rod overheating without pumping in large quantities of water which releases radioactive pollution into the environment via steam and water runoff.  An immense amount of radioactive material has already been released into the atmosphere affecting a vast area of Japan as well as the ocean.   Many workers at the plant are expected to die quickly due to radiation exposure.  We don’t know how and when the continuing release of radiation can be contained.  Much of Japan including Tokyo could be contaminated semi-permanently by fallout resulting in radiation levels orders of magnitude higher than are normally acceptable.  Tens of millions of people are likely to be effected. 

An excellent source of information on the situation in Japan is the blog All Things Nuclear.  The March 30 entry relates:Today the IAEA has finally confirmed what some analysts have suspected for days: that the concentration per area of long-lived cesium-137 (Cs-137) is extremely high as far as tens of kilometers from the release site at Fukushima Dai-Ichi, and in fact would trigger compulsory evacuation under IAEA guidelines. — The IAEA is reporting that measured soil concentrations of Cs-137 as far away as Iitate Village, 40 kilometers northwest of Fukushima-Dai-Ichi, correspond to deposition levels of up to 3.7 megabecquerels per square meter (MBq/sq. m). This is far higher than previous IAEA reports of values of Cs-137 deposition, and comparable to the total beta-gamma measurements reported previously by IAEA and mentioned on this blog. — This should be compared with the deposition level that triggered compulsory relocation in the aftermath of the Chernobyl accident: the level set in 1990 by the Soviet Union was 1.48 MBq/sq. m.  — Thus, it is now abundantly clear that Japanese authorities were negligent in restricting the emergency evacuation zone to only 20 kilometers from the release site.”  Other blog entries refer to the release of substantial quantities of radioactive iodine-131 and to how plutonium is being found in soil samples.

Background on nuclear radiation

For the general Japanese public, the biggest concern is not direct radiation from the Fukushima plant but rather radioactive substances which are released into the environment either via the atmosphere or via runoff into the ocean.  Radioactive substances can enter the body via the air, groundwater, tap water or foods or vegetables consumed. Three radioactive substances of primary concern are:

Radio iodine-131:  Although having a half life of only 8 days, iodine-131 can be dangerous if ingested by someone with a thyroid iodine deficiency.  It is a beta radiation emitter, which means the radiation is not a threat unless the emitting substance is inside the body. “Due to its mode of beta decay, iodine-131 is notable for causing mutation and death in cells which it penetrates, and other cells up to several millimeters away. For this reason, high doses of the isotope are sometimes paradoxically less dangerous than low doses, since they tend to kill thyroid tissues which would otherwise become cancerous as a result of the radiation. For example, children treated with moderate dose of I-131 for thyroid adenomas had a detectable increase in thyroid cancer, but children treated with a much higher dose did not(ref).”  As long as the damaged Fukushima plant continues to release iodine-131 into the environment, it will be a threat. 

Caesium-137: Has a half life of about 30 years and emits beta and gamma radiation.  Because it is chemically similar to potassium, it is readily absorbed in vegetables and human bodies.  “Caesium-137 is water-soluble, and the biological behavior of caesium is similar to that of potassium and rubidium. After entering the body, caesium gets more or less uniformly distributed throughout the body, with higher concentration in muscle tissues and lower in bones. The biological half-life of caesium is rather short at about 70 days.[6] Experiments with dogs showed that a single dose of 3800 μCi/kg (approx. 44 μg/kg of caesium-137) is lethal within three weeks[7](ref).”

Plutonium:  Plutonium is a byproduct of nuclear reactions, is the heaviest stable element and has been called ‘the most toxic substance known to man,” emitting alpha, beta and gamma radiation.  The most important isotope of plutonium is plutonium-239, with a half-life of 24,100 years..  Plutonium-238 has a half-life of 88 years and emits alpha particles. plutonium-244 has a half-life of about 80 million years. “Isotopes and compounds of plutonium are radioactive poisons that accumulate in bone marrow. Contamination by plutonium oxide (spontaneously oxidized plutonium) has resulted from a number of nuclear disasters and radioactive incidents including military nuclear accidents where nuclear weapons have burned.[86] Studies of the effects of these smaller releases, as well as of the widespread radiation poisoning sickness and death following the Atomic bombings of Hiroshima and Nagasaki, have provided considerable information regarding the dangers, symptoms and prognosis of radioactive poisoning. PMID 19454804(ref).”

As reported in the news, higher than normal levels of both iodine-131 and caesium-137 have been noted not only in Japan but worldwide due to the Fukushima incident.  And plutonium has been found in soil samples near the reactor sites.

According to the International Atomic Energy Agency Briefing on Fukushima Nuclear Accident, 31 March 2011 (14:00 UTC), On 30 March, deposition of iodine-131 was detected in 8 prefectures, and deposition of cesium-137 in 12 prefectures. On 30 March in the prefectures where deposition of iodine-131 was reported, the range was from 2.5 to 240 becquerel per square metre. For caesium-137, the range was from 3 to 57 becquerel per square metre. In the Shinjyuku district of Tokyo, the daily deposition of both iodine-131 and cesium-137 on 30 March was below 30 becquerel per square metre. — Since our briefing of yesterday, significant data related to food contamination has been submitted by the Japanese Ministry of Health, Labour and Welfare. Seventy-six samples were taken from 28-30 March, and reported on 30 March. Analytical results for 51 of the 76 samples for various vegetables, fruit (strawberry), seafood (sardines), and unprocessed raw milk in eight prefectures (Chiba, Fukushima, Gunma, Ibaraki, Kanagawa, Niigata, Saitama, and Yamagata), indicated that iodine-131, caesium-134 and caesium-137 were either not detected or were below the regulation values set by the Japanese authorities. However, it was reported that analytical results in Fukushima prefecture for the remaining 25 of the 76 samples for broccoli, cabbage, rapeseed, spinach and other leafy vegetables, indicated that iodine-131 and/or caesium-134 and caesium-137 exceeded the regulation values set by the Japanese authorities.”

Right now, as reported in the news, the biggest dangers from iodine-131 and caesium-137 due to the Fukushima situation are within a 100 kilometer distance of the power plant site, although levels significantly higher than normal have been reported elsewhere in Japan. Higher levels of these substances due to the Fukushima situation have also been observed in the US, both on the West and East coasts.  These US levels are so-far significantly below those considered to offer health risks.  As the damaged reactors and stored fuel rods continue to release large quantities of the radioactive substances, and because major new releases due to possible meltdowns could occur, however, the danger exists that the relatively comfortable situation in the US could change. And there is the danger of consuming contaminated vegetables or seafood anywhere it may be shipped to.   

Biological impact from radiation damage due to ingested substances is different than radiation from sources outside the body in several important respects including:  1.  Alpha and beta radiation is very short range so is of no threat when coming from substances outside the body unless those substances are touched, inhaled, eaten or otherwise penetrate the body and 2.  Radiation from a substance like iodine-131 or caesium-137 that is absorbed in body tissues continues to create damage as long as that substance is within the body.

Consequences of radiation damage

Radiation damage can be short term or long term and is measured by exposure.  Radiation dosages are measured in sieverts.  The sievert (symbol: Sv) is the SI derived unit of dose equivalent radiation. It attempts to quantitatively evaluate the biological effects of ionizing radiation as opposed to the physical aspects, which are characterised by the absorbed dose, measured in gray. It is named after Rolf Sievert, a Swedish medical physicist renowned for work on radiation dosage measurement and research into the biological effects of radiation(ref).”

This link contains a table relating exposure in sieverts to biological impacts.  Annual limit on intake (ALI) is the derived limit for the amount of radioactive material taken into the body of an adult worker by inhalation or ingestion in a year. ALI is the intake of a given radionuclide in a year that would result in:

  • a committed effective dose equivalent of 0.05 Sv (5 rems) for a “reference human body”, or
  • a committed dose equivalent of 0.5 Sv (50 rems) to any individual organ or tissue,

whatever dose is the smaller[8](ref).”  The average natural radiation dose is 2.4 mSv per year, with a “typical range” reaching up to 10 mSv.  As the table shows, a short-term dose of as little as 2 sieverts can lead to severe medical problems and a 5-50% chance of death even with medical care.  And any dose of over 8 sieverts is sure to lead to rapid death.

The CDC has prepared a fact sheet Acute Radiation Syndrome (ARS): A Fact Sheet for the Public. 

Longer-term exposure to even low intensities of radiation can lead to mutations of cell-cycle genes and cancers

Protective measures

Known protective measures can be taken when ingestion of one of the mentioned substances is unavoidable or has already happened.  These are time-honored approaches known for decades.  Specifically:

Potassium iodide for iodine-131

Absorption of iodine-131 can be significantly reduced or avoided by taking potassium iodide, though taking such pills after exposure to iodine-131 may be useless.  A recent article in Pharmacy Practice News reports “As alarmed residents visit various Internet sites to purchase what is touted as a panacea for radiation sickness, costs of KI have skyrocketed. According to some reports, a package of 14 tablets that normally costs about $10 has soared to $540 in some ebay.com listings—a shocking 5,300% increase. Although some manufacturers are struggling to meet demands for KI in this suddenly lucrative market, others have simply given up, temporarily ceasing production.1-4  And this increase is in the US, not Japan.  Further, the article reports “In situations where internal radiation contamination is suspected, the initial concern is inhalation of radioactive iodine, which is readily absorbed by the thyroid gland, leading to thyroid injury/cancer, particularly in infants/fetus, children and patients with low iodine stores. KI is known to flood the thyroid gland with stable iodine, preventing the organ from absorbing radioactive iodine—but it can only go so far. Consumers should be told that KI only protects the thyroid (not other parts of the body) from radioactive iodine—it cannot reverse effects of radioactive iodine once damage to the thyroid has occurred—and it cannot protect the body from radioactive elements other than radioactive iodine.5 — A dose of KI is recommended before or immediately following radiation exposure in all children and adults under the age of 40 years who are not allergic to iodine or shellfish, and if radiation still poses a threat 24 hours later, a repeat dose is administered (unless the patient is pregnant or breastfeeding, or a newborn infant).”  Use of potassium iodide in other than emergency situations is definitely not recommended.  You can see the document Prophylactic Use of Potassium Iodide (KI) in Radiological Emergencies – Information for Physicians. 

 

Chelating Plutonium via DPTA

The 2005 publication Chelating agents used for plutonium and uranium removal in radiation emergency medicine points to an approach for removal of ingested plutonium.  A chelating agent is one that chemically grabs molecules of an offending substance for transportation outside of the body.  DPTA is a chelating agent that is approved by the FDA for removal of plutonium as well as americium, and curium which are also radioactive.  The CDC document Facts About DTPA provides some basic information about the substance including: “DTPA is a kind of medicine called a chelating agent. Chelating agents work by binding and holding on to radioactive materials or poisons that get into the body. Once bound to a radioactive material or poison, the chelating agent is then passed from the body in the urine. Chelating agents help decrease the amount of time it takes to get a poison out of the body. — What does DTPA do?  When radioactive materials get into the body through breathing, eating, drinking, or through open wounds, we say that “internal contamination” has occurred. Over the past 50 years, almost all cases of internal contamination have happened in people who use radioactive materials in their work. Since the 1960s, doctors have used DTPA as a chelating agent to treat internal contamination from radioactive materials such as americium, plutonium, californium, curium, and berkelium. Currently, DTPA is approved by the U.S. Food and Drug Administration (FDA) for chelation of only three radioactive materials: plutonium, americium, and curium. — What DTPA cannot do: Knowing what DTPA cannot do is also important. DTPA cannot bind all of the radioactive materials that might get into a person’s body after a radiological or nuclear event, such as a terrorist attack with a “dirty bomb.” This medicine cannot prevent radioactive materials from entering the body. DTPA cannot reverse the health effects caused by radioactive materials once these materials have entered the body. — How does DTPA work?  DTPA comes in two forms: calcium (Ca-DTPA) and zinc (Zn-DTPA). Both forms work by tightly chelating (holding on to) plutonium, americium, and curium. These radioactive materials (bound to DTPA) are then passed from the body in the urine. When given within the first day after internal contamination has occurred, Ca-DTPA is about 10 times more effective than Zn-DTPA at chelating plutonium, americium, and curium. After 24 hours have passed, Ca-DTPA and Zn-DTPA are equally effective in chelating these radioactive materials. — How well does DTPA work?  Chelating agents work best when given shortly after radioactive materials or poisons have entered the body. The more quickly a radioactive material or poison is removed from the body, the fewer and less serious the health effects will be. After 24 hours, plutonium, americium, and curium are harder to chelate. However, DTPA can still work to remove these radioactive materials from the body several days or even weeks after a person has been internally contaminated. — Who should get DTPA? Many people could be internally contaminated after a radiological or nuclear terrorist event. People contaminated with small amounts of radioactive materials might not need treatment with DTPA. Doctors and public health authorities will work together to decide who will likely benefit from DTPA treatment.”  The literature related to plutonium chelation tends to be old with some articles going back 50 years or more(ref).

Clearing Caesium-137 with Prussian Blue

The CDC fact sheet Prussian blue tells the story: “Prussian blue can remove certain radioactive materials from people’s bodies, but must be taken under the guidance of a doctor. — People may become internally contaminated (inside their bodies) with radioactive materials by accidentally ingesting (eating or drinking) or inhaling (breathing) them, or through direct contact (open wounds). The sooner these materials are removed from the body, the fewer and less severe the health effects of the contamination will be. Prussian blue is a substance that can help remove certain radioactive materials from people’s bodies. However, small amounts of contamination may not require treatment. Doctors can prescribe Prussian blue if they determine that a person who is internally contaminated would benefit from treatment. — What Prussian blue is:  Prussian blue was first produced as a blue dye in 1704 and has been used by artists and manufacturers ever since. It got its name from its use as a dye for Prussian military uniforms. Prussian blue dye and paint are still available today from art supply stores. — People SHOULD NOT take Prussian blue artist’s dye in an attempt to treat themselves. This type of Prussian blue is not designed to treat radioactive contamination and is not made for that purpose. People who are concerned about the possibility of being contaminated with radioactive materials should go to their doctors for advice and treatment.  Use of Prussian blue to treat radioactive contamination: Since the 1960s, Prussian blue has been used to treat people who have been internally contaminated with radioactive cesium (mainly Cs-137) and nonradioactive thallium (once an ingredient in rat poisons). Doctors can prescribe Prussian blue at any point after they have determined that a person who is internally contaminated would benefit from treatment. Prussian blue will help speed up the removal of cesium and thallium from the body. —How Prussian blue works: Prussian blue traps radioactive cesium and thallium (mainly Tl-201) in the intestines and keeps them from being re-absorbed by the body. The radioactive materials then move through the intestines and are excreted (passed) in bowel movements. Prussian blue reduces the biological half-life1 of cesium from about 110 days to about 30 days. Prussian blue reduces the biological half-life of thallium from about 8 days to about 3 days. Because Prussian blue reduces the time that radioactive cesium and thallium stay in the body, it helps limit the amount of time the body is exposed to radiation. — Who can take Prussian blue/ The drug is safe for most adults, including pregnant women, and children (2 ─12 years). Dosing for infants (ages 0 ─2 years) has not been determined yet. Women who are breast feeding their babies should stop breast feeding if they think they are contaminated with radioactive materials and consult with their doctors. People who have had constipation, blockages in the intestines, or certain stomach problems should be sure to tell their doctors before taking Prussian blue. Before taking Prussian blue, people also should be sure to tell their doctors about any other medicine they are taking. — How Prussian blue is given:  Prussian blue is given in 500-milligram capsules that can be swallowed whole. People who cannot swallow pills can take Prussian blue by breaking the capsules and mixing the contents in food or liquid. Breaking open the capsules will cause people’s mouths and teeth to be blue during the time of treatment. — The dose of Prussian blue depends on the person’s age and the amount of contamination in the body. Prussian blue usually is given 3 times a day for a minimum of 30 days, depending on the extent of the contamination. – Warning:  People SHOULD NOT take Prussian blue artist’s dye in an attempt to treat themselves. This type of Prussian blue is not designed to treat radioactive contamination and is not made for that purpose. People who are concerned about the possibility of being contaminated with radioactive materials should go to their doctors for advice and treatment.”

Taking antioxidants to minimize radiation damage

The above treatments are directed to elimination of dangerous radioactive substances from the body and may constitute a first level of defense in case of ingestion of radioactive substances through breathing, eating, drinking or exposure of open wounds.  As recommended by the CDC they should be undertaken only under the guidance of a physician.  Even using these approaches, however, the various radioactive substances will be resident in the body for a certain period and can possibly create serious damage depending on exposure dose and how quickly the removal action is initiated.  There is a second level and complimentary of defense possible, and that is to minimize the damaging biological impact of radiation through taking a regimen of antioxidants.

I have written about the use of antioxidants to protect against the negative effects of medical imaging radiation first in a 2008 position paper Protection Against Radiation – The Second Line of Defense, and also briefly in an August 2009 blog entry Medical radiation risk – you can do something about it.  Medical imaging radiation usually originates from outside the body and is electromagnetic in nature making it akin to gamma radiation such as is generated by an X-ray machine or as exists in outer space.  Many studies have shown that antioxidants can be protective against biological damage caused by such exterior-originated radiation.  Many such studies are cited in my position paper and a few new ones are cited here below.  Much of this work has been sponsored by NASA and has been concerned with safety in space of astronauts. 

Would antioxidants also provide protection from in-body radiation sources such as iodine-131 or caesium-137?  The mechanisms of radiation damage are the same.  Ionizing radiation, whatever the source may be, knocks electrons loose from molecules producing free radicals.  These free radicals can propagate producing secondary showers of free radicals, particularly when generated by high-energy photons from gamma radiation.  Free radicals can generate multiple forms of damage including to tissue structures, body cell DNA, germ-line DNA and mitochondrial DNA.  From my treatise on radiation and antioxidants: “Ionizing x-radiation in any quantities is potentially deleterious to health. Radiation ionizes oxygen to produce Reactive Oxygen Species (ROS) like OH which steal electrons from lipids in cell membranes, a process called lipid peroxidation. A chain of damaging events can be let loose from a single high-energy electron event as unstable fatty acid radicals propagating in tissues produce other unstable radicals. The result can be damage to DNA or mitochondrial DNA, mangled chromosomes, protein cross-linking, cell apoptosis, genetic mutations, mutated germ cells and other forms of cell havoc. Radiation damage can show up in many ways including skin erythema, hair loss, vascular damage, internal bleeding, cataracts, cancers, weakened immune systems, sterility, mutations in offspring, premature ageing and death. Cell DNA repair mechanisms are effective in correcting some radiation-induced damage but may themselves be compromised by radiation.”

Newer literature references re antioxidants for radiation protection

My treatise PROTECTION AGAINST RADIATION – – THE SECOND LINE OF DEFENSE, last updated in 2008, provides a thorough discussion of how antioxidants are radioprotective and cites 30 of some 110 applicable literature citations I came across up to that point. Also you can check on the 2008 publication Antioxidants Reduce Consequences of Radiation Exposure.  The following are selected updates on relevant research as reported in Science Daily articles.

A March 2011 Science Daily story Antioxidant Formula Prior to Radiation Exposure May Prevent DNA Injury, Trial Suggests reports: “A unique formulation of antioxidants taken orally before imaging with ionizing radiation minimizes cell damage, noted researchers at the Society of Interventional Radiology’s 36th Annual Scientific Meeting in Chicago, Ill. In what the researchers say is the first clinical trial of its kind, as much as a 50 percent reduction in DNA injury was observed after administering the formula prior to CT scans. — People are 70 percent water, and X-rays collide with water molecules to produce free radicals (groups of atoms with an unpaired number of electrons that are dangerous when they react with cellular components, causing damage and even cell death) that can go on to do damage by direct ionization of DNA and other cellular targets, noted Murphy. The research team evaluated whether a special combination of antioxidants have an ability to neutralize these free radicals before they can do damage. — “Our intent was to develop an effective proprietary formula of antioxidants to be taken orally prior to exposure that can protect a patient’s DNA against free radical mediated radiation injury, and we have applied to patent this formulation and a specific dose strategy,” said Murphy. — The experiments measured DNA damage as a surrogate marker for DNA injury.”

A September 2008 Science Daily story Plant Antioxidant May Protect Against Radiation Exposure reports on research specifically concerned with identifying protective measures in case of a nuclear emergency: “Resveratrol, the natural antioxidant commonly found in red wine and many plants, may offer protection against radiation exposure, according to a study by the University of Pittsburgh School of Medicine. When altered with acetyl, resveratrol administered before radiation exposure proved to protect cells from radiation in mouse models.”  — The study, led by Joel Greenberger, M.D., professor and chairman of the Department of Radiation Oncology at the University of Pittsburgh School of Medicine, is overseen by Pitt’s Center for Medical Countermeasures Against Radiation. The center is dedicated to identifying and developing small molecule radiation protectors and mitigators that easily can be accessed and administered in the event of a large-scale radiological or nuclear emergency. — “New, small molecules with radioprotective capacity will be required for treatment in case of radiation spills or even as countermeasures against radiological terrorism,” said Dr. Greenberger. “Small molecules which can be easily stored, transported and administered are optimal for this, and so far acetylated resveratrol fits these requirements well.” — “Currently there are no drugs on the market that protect against or counteract radiation exposure,” he added. “Our goal is to develop treatments for the general population that are effective and non-toxic. — Dr. Greenberger and his team are conducting further studies to determine whether acetylated resveratrol eventually can be translated into clinical use as a radioprotective agent. In 2004, this same team of researchers identified the drug JP4-039, which can be delivered directly to the mitochondria, the energy producing areas of cells. When this occurs, the drug assists the mitochondria in combating radiation-induced cell death. — The results of the research were presented during the American Society for Therapeutic Radiology and Oncology’s (ASTRO) 50th Annual Meeting in Boston.”

An October 2008 2009 story Herbal Tonic For Radiotherapy? Gingko Biloba Tree May Protect Cells From Radiation Damage reports on radioprotection provided by Gingko biloba against gamma radiation from caesium-137: ” Antioxidant extracts of the leaves of the Gingko biloba tree may protect cells from radiation damage, according to a study published in the International Journal of Low Radiation. — Chang-Mo Kang of the Korea Institute of Radiological and Medical Sciences in Taegu and colleagues are interested in the protective effects of well-known herbal remedies of which Gingko biloba is one. G. biloba is a unique tree species with no close living relatives and extracts of its leaves contain antioxidant compounds including glycosides and terpenoids known as ginkgolides and bilobalides. — These compounds are thought to protect cells from damage by free radicals and other reactive oxidizing species found in the body. These are generated continuously by the body’s normal metabolism, and in excess in some diseases or after exposure to pollution or radiation. They damage proteins, DNA and other biomolecules and left unchecked can kill cells. — As such, extracts of certain plants that contain antioxidants, including G. biloba, have attracted interest for their pharmacological activity. G. biloba is currently sold as a herbal supplement and there are numerous claims for health benefits, including the possibility of preventing the onset of dementia or Alzheimer’s disease. — Kang and colleagues have now collected human white blood cells, lymphocytes, from healthy donors aged 18 to 50 years. They treated half of these cells with commercially available G. biloba extract in the laboratory and doused the other half with salt solution as an experimental control. They then compared the effects of gamma radiation from radioactive cesium on the white blood cells compared to the untreated control samples. — The team uses a light microscope to look for lymphocytes undergoing programmed cell death, or apoptosis, as a result of radiation exposure. They found that there was a significant increase in apoptosis in the untreated cells compared with those treated with G. biloba extract. Almost a third of the untreated cells underwent apoptosis compared with approximately one in twenty of the treated cells. Parallel studies with laboratory mice also demonstrated a similar protective effect against radiation poisoning. — The results suggest that the extracts can neutralize the free-radicals and oxidizing agents produced in the cells by the radiation and so prevent them from undergoing apoptosis.”

The July 2009 Science Daily story New Pill May Prevent Injury After Radiation Exposure reports “Researchers from Boston University School of Medicine (BUSM) and collaborators have discovered and analyzed several new compounds, collectively called the ”EUK-400 series,” which could someday be used to prevent radiation-induced injuries to kidneys, lungs, skin, intestinal tract and brains of radiological terrorism victims. The findings, which appear in the June issue of the Journal of Biological Inorganic Chemistry, describe new agents which can be given orally in pill form, which would more expedient in an emergency situation. — These agents are novel synthetic “antioxidants” that protect tissues against the kind of damage caused by agents such as “free radicals.” Free radicals, and similar toxic byproducts formed in the body, are implicated in many different types of tissue injury, including those caused by radiation exposure. Often, this kind of injury occurs months to years after radiation exposure. The BUSM researchers and their colleagues are developing agents that prevent injury even when given after the radiation exposure. — This paper describes a newer class of compounds, the ”EUK-400 series,” that are designed to be given as a pill. According to the researchers, experiments described in their paper prove that these agents are orally active. They also show that the new agents have several desirable “antioxidant” activities, and protect cells in a “cell death” model. — These same BUSM researchers and collaborators had previously discovered novel synthetic antioxidants that effectively mitigate radiation injuries, but had to be given by injection. “We have developed some of these agents and have studied them for over 15 years beginning with our work at the local biotechnology company Eukarion,” said senior author Susan Doctrow, PhD, a research associate professor of medicine at BUSM’s Pulmonary Center. “These injectible antioxidants are very effective, but there has also been a desire to have agents that can be given orally. A pill would be more feasible than an injection to treat large numbers of people in an emergency scenario,” she adds.”

The 2007 article Antioxidants Could Provide All-Purpose Radiation Protection reports “Two common dietary molecules found in legumes and bran could protect DNA from the harmful effects of radiation, researchers from the University of Maryland report. Inositol and inositol hexaphosphate (IP6) protected both human skin cells and a skin cancer-prone mouse from exposure to ultraviolet B (UVB) radiation, the damaging radiation found in sunlight, the team reported November 5 at the American Association for Cancer Research Centennial Conference on Translational Cancer Medicine. — According to the researchers, inositol and IP6 could decrease the severity of side effects from radiation therapy, saving healthy cells while simultaneously increasing the potency of the treatment against cancer cells. Both molecules are potent antioxidants, the Maryland researchers say, capable of preventing reactive molecules from injuring DNA and turning cells cancerous. — “Both of these potent antioxidants have been shown to have broad-spectrum anti-tumor capabilities, and now our studies confirm the degree to which these molecules protect against the DNA-damaging effects of ionizing radiation,” said Abulkalam M. Shamsuddin, M.D., professor of pathology at the University of Maryland School of Medicine. “Radiation damage is radiation damage, regardless of the source, so there could also be a protective role for IP6 in any form of radiation exposure, whether it is from a therapeutic dose or from solar, cosmic or nuclear sources.” – Normally, cells permanently damaged by radiation undergo a genetically programmed process of cell suicide, called apoptosis. Shamsuddin reports that UVB-irradiated human keratinocytes, when treated with IP6, were more likely to survive. Untreated skin cells were more likely to undergo apoptosis, indicating that the DNA in those cells was damaged irreparably and fatally. According to Shamsuddin, the treated cells take an extended pause at the point in the cellular life cycle where innate mechanisms repair DNA before the cell divides. — “IP6 certainly has some interactivity with DNA, but how exactly it works to repair DNA is still something of a mystery. There are reports that IP6 binds with DNA repair molecule Ku to bring about the repair process,” Shamsuddin said. — According to Shamsuddin, IP6 could also offer protection against accidents or purposeful incidents involving nuclear material. “It could also be advisable to use IP6 plus inositol as a cautionary treatment following a nuclear disaster or dirty bomb,” Shamsuddin said.”

Drugs under development that mitigate radiation damage

The April 2009 story Developmental Drug Helps Protect Against Radiation Damage reports “The study, led by Joel Greenberger, M.D., professor and chairman of the Department of Radiation Oncology at Pitt, is overseen by Pitt’s Center for Medical Countermeasures Against Radiation. The center is dedicated to identifying and developing small molecule radiation protectors and mitigators that can be easily accessed and administered in the event of a large-scale radiological or nuclear emergency. — JP4-039 assists the mitochondria, the energy generator of all cells, in combating irradiation-induced cell death. For this study, cells treated immediately after irradiation with JP4-039 demonstrated significant radioprotection, suggesting a potential role for the drug as a mitigator of radiation damage. — “Currently, no drugs on the market counteract the effects of radiation exposure,” said Dr. Greenberger. “We know this drug can counteract the damage caused by irradiation, and now we want to develop the ideal dosage, one that is effective for the general population while remaining non-toxic. Our goal is to take this drug through a phase I clinical trial and, once the dosage is established, develop the drug for late-stage clinical trials and market licensing.” —  The study, led by Joel Greenberger, M.D., professor and chairman of the Department of Radiation Oncology at Pitt, is overseen by Pitt’s Center for Medical Countermeasures Against Radiation. The center is dedicated to identifying and developing small molecule radiation protectors and mitigators that can be easily accessed and administered in the event of a large-scale radiological or nuclear emergency. — JP4-039 assists the mitochondria, the energy generator of all cells, in combating irradiation-induced cell death. For this study, cells treated immediately after irradiation with JP4-039 demonstrated significant radioprotection, suggesting a potential role for the drug as a mitigator of radiation damage. — “Currently, no drugs on the market counteract the effects of radiation exposure,” said Dr. Greenberger. “We know this drug can counteract the damage caused by irradiation, and now we want to develop the ideal dosage, one that is effective for the general population while remaining non-toxic. Our goal is to take this drug through a phase I clinical trial and, once the dosage is established, develop the drug for late-stage clinical trials and market licensing.”

The 2008 story Could a Nanotube-Based Drug Prevent Radiation Injury? reports “The Department of Defense has commissioned a nine-month study from Rice University chemists and scientists in the Texas Medical Center to determine whether a new drug based on carbon nanotubes can help prevent people from dying of acute radiation injury following radiation exposure. The new study was commissioned after preliminary tests found the drug was greater than 5,000 times more effective at reducing the effects of acute radiation injury than the most effective drugs currently available. — “More than half of those who suffer acute radiation injury die within 30 days, not from the initial radioactive particles themselves but from the devastation they cause in the immune system, the gastrointestinal tract and other parts of the body,” said James Tour, Rice’s Chao Professor of Chemistry, director of Rice’s Carbon Nanotechnology Laboratory (CNL) and principal investigator on the grant. “Ideally, we’d like to develop a drug that can be administered within 12 hours of exposure and prevent deaths from what are currently fatal exposure doses of ionizing radiation.” — The drug is based on single-walled carbon nanotubes, hollow cylinders of pure carbon that are about as wide as a strand of DNA. To form NTH, Rice scientists coat nanotubes with two common food preservatives — the antioxidant compounds butylated hydroxyanisole (BHA) and butylated hydroxytoluene (BHT) — and derivatives of those compounds. — “The same properties that make BHA and BHT good food preservatives, namely their ability to scavenge free radicals, also make them good candidates for mitigating the biological affects that are induced through the initial ionizing radiation event,” Tour said. — In preliminary tests at M.D. Anderson in July 2007, mice showed enhanced protection when exposed to lethal doses of ionizing radiation when they were given first-generation NTH drugs prior to exposure.”

Wrapping it up

1.     By far, the best protection against nuclear radiation is avoiding it.

2.     We do not know when the release of radioactive substances into the environment from the earthquake and tsunami-struck Fukushima Daiichi nuclear power plant in Japan will stop.  So far, efforts to bring the situation under control have not succeeded.

3.     Two radioactive substances of particular concern are iodine-131 and caesium-137.  These have already been released into the environment in large quantities due to the Fukushima disaster.  These substances are both highly radioactive and highly bioactive, being readily absorbed into human bodies.  Also of serious concern is plutonium, an extremely toxic byproduct of nuclear fission that has been found in the ground near the reactor sites. 

4.     Consequences from internal exposure to these substances can be most serious both in the short and long term.  Each of these substances in critical doses can cause death if left lodged in the body.

5.     Radiation from ingested radioactive substances originating from a nuclear accident is in some respects similar to radiation received from external sources, in some respects different.  The big dangers from of iodine-131, caesium-137 or plutonium come into play if they are absorbed into the body via breathing, eating or drinking them.  Caesium-137, for example, is readily absorbed from soil into certain vegetables and can enter the body effectively by eating such contaminated vegetables.

6.     Traditional means exist for removing these substances from bodies or preventing their absorption including use of potassium iodide, Prussian blue, and  DPTA.  These approaches should be pursued only under medical supervision.

7.     Effectively removing or preventing adsorption of iodine-131, caesium-137 or plutonium using these approaches takes time, may be only partially effective, and may still leave the result of substantial radiation injury under the best of circumstances.

8.     Antioxidants have been shown to be at least partially effective in reducing the damaging effects of radiation from external sources.  The known mechanisms of radiation damage suggests that the same should be the case for radiation from ingested sources.

9.     Research studies have established that a number of conventional antioxidants are protective against internal radiation damage including but not limited to resveratrol, alpha-lipoic acid, ginko biloba, beta carotene, curcumin, inositol, selenium, melatonin, vitamin C, vitamin E, N-acetylcysteine and ginseng(ref)(ref)(ref).

10.                        A number of new antioxidant and antioxidant combinations as well as other substances are being investigated for their superior abilities to protect against radiation.  How their effectiveness compares against that of conventional antioxidants is unknown. 

Posted in Uncategorized | 4 Comments

The epigenetic regulation of telomeres

This blog entry focuses on a specific aspect of telomere biology, the epigenetic regulation of telomeres as mediated by TERRA, a recently-discovered RNA. Most popular discussions related to telomeres and telomerase are still based on simplistic earlier models of telomerase biology which do not take TERRA into account. So are the increasingly-popular commercial approaches to telomere lengthening via “telomerase activator” substances. The new TERRA-related research may explain how diet, certain supplements and lifestyle factors like exercise so profoundly affect telomere lengths and can inhibit cell senescence.
First, as a preamble or appetizer for what follows, regarding the relationship of telomere length and aging I invite you to view this 3-minute video.

Background on telomeres and telomerase.
Although this blog entry focuses on epigenetic regulation as a new and very important aspect of telomere biology, I continue to stand behind what I have written related to telomeres and telomerase reflecting a shift over a three-year period. The most-recent relevant blog entries were written in October 2010: Telomere lengths, Part 3: Selected current research on telomere-related signaling, telomere lengths, cancers and disease processes, Part2: lifestyle, dietary, and other factors associated with telomere shortening and lengthening, and Part1: telomere lengths, cancers and disease processes. These entries contain a great deal of information as well as links to multiple earlier blog entries on telomeres and telomerase. And, of course, the 12th theory of aging discussed in my treatise is Telomere Shortening and Damage. Three years ago, I thought that taking astragaloside IV or cycloastragenol supplements to extend telomeres was possibly a good anti-aging intervention. I no longer see that as the case.
In the abovementioned Part 3 blog entry I discussed how damaged or too-shortened telomeres can lead to epigenetic changes. “Another important contribution to the picture of how cell senescence affects aging is provided by the October 2010 publication Reduced histone biosynthesis and chromatin changes arising from a damage signal at telomeres.
During replicative aging of primary cells morphological transformations occur, the expression pattern is altered and chromatin changes globally. Here we show that chronic damage signals, probably caused by telomere processing, affect expression of histones and lead to their depletion. We investigated the abundance and cell cycle expression of histones and histone chaperones and found defects in histone biosynthesis during replicative aging. Simultaneously, epigenetic marks were redistributed across the phases of the cell cycle and the DNA damage response (DDR) machinery was activated. The age-dependent reprogramming affected telomeric chromatin itself, which was progressively destabilized, leading to a boost of the telomere-associated DDR with each successive cell cycle.” — This same publication puts forward an important new concept: “We propose a mechanism in which changes in the structural and epigenetic integrity of telomeres affect core histones and their chaperones, enforcing a self-perpetuating pathway of global epigenetic changes that ultimately leads to senescence.” Histones, we recall, are the spindles around which DNA is wrapped, and chaperones are “are proteins that assist the non-covalent folding or unfolding and the assembly or disassembly of other macromolecular structures.” in this case the chaperones are ones that assist folding of DNA in histone structures, HSP90 being a main one discussed below.”
Rather than discussing how shortened or damaged telomeres lead to epigenetic changes, the present blog entry is focused on how epigenetic changes arising from other causes (dietary, lifestyle, aging, etc.) can affect telomere health, regulate telomere lengths, and impinge on cell senescence.
About telomere structures, shelterin and telomere binding proteins
Telomeres are commonly described as simple repeats of inert DNA at the ends of chromosomes, like the tips of shoelaces. In fact, they are quite complex structures. The 2011 publication Healthy aging and disease: role for telomere biology? Contains the following description and diagrams: “Telomeres are specialized DNA structures located at the terminal ends of the chromosomes [5]. Their primary function is to maintain genome stability. Telomeres consist of non-coding double-stranded repeats of guanine-rich tandem DNA sequences (TTAGGG) that are extended 9–15 kb in humans and end in a 50–300 nucleotide 3′ single guanine strand overhang [6]. This overhang can fold back and invade the double-stranded telomere helix, forming a large ‘T-loop’. The T-loop facilitates the formation of a high-order structure mediating the end-capping [7] (Figure 1) (ref).”
“The stability of the T-loop is largely dependent on the integrity of the associated telomere-specific proteins, also called the shelterin complex [8] (Figure 2). The proteins involved in the shelterin complex include TRF (telomeric repeat-binding factor) 1 and TRF2, which bind to the double-stranded segment of telomeric DNA [9,10] (ref).”
“POT1 (protein protection of telomeres 1) binds directly to the single-stranded telomeric DNA and interacts directly with TPP1 (tripeptidyl peptidase 1) [11]. Rap1 (repressor activator protein 1) binds TRF2 [12], and TIN2 (TRF1-interacting nuclear factor 2) is a central component of the complex interacting with TRF1, TRF2 and TPP1 [13,14]. The telomere complex plays a critical role by protecting the chromosomes from recognition by the DNA damage-repair system as DNA ‘breaks’ and activation of the p53 or p16INK4a pathway, eventually leading to cellular senescence or apoptosis [15](ref).”
Since these diagrams don’t show TERRA, the actual situation is even more complex. TERRA accompanies the telomeric DNA binding to TRF2.

The 2009 publication TERRA: telomeric repeat-containing RNA provides additional detail: “Telomeres, the physical ends of eukaryotic chromosomes, consist of tandem arrays of short DNA repeats and a large set of specialized proteins. A recent analysis has identified telomeric repeat-containing RNA (TERRA), a large non-coding RNA in animals and fungi, which forms an integral component of telomeric heterochromatin. — TERRA transcription occurs at most or all chromosome ends and it is regulated by RNA surveillance factors and in response to changes in telomere length. TERRA functions that are emerging suggest important roles in the regulation of telomerase and in orchestrating chromatin remodelling throughout development and cellular differentiation. — The accumulation of TERRA at telomeres can also interfere with telomere replication, leading to a sudden loss of telomere tracts. Such a phenotype can be observed upon impairment of the RNA surveillance machinery or in cells from ICF (Immunodeficiency, Centromeric region instability, Facial anomalies) patients, in which TERRA is upregulated because of DNA methylation defects in the subtelomeric region. Thus, TERRA may mediate several crucial functions at the telomeres, a region of the genome that had been considered to be transcriptionally silent.”

While the role of TERRA in regulation of telomeres is central to this blog entry, I have referred to it in the Part 3 post mentioned above. The 2009 publication TERRA RNA Binding to TRF2 Facilitates Heterochromatin Formation and ORC Recruitment at Telomeres is one of several recent publications illustrating the wheels-within-wheels complexity of telomere formation and maintenance. “Telomere-repeat-encoding RNA (referred to as TERRA) has been identified as a potential component of yeast and mammalian telomeres. We show here that TERRA RNA interacts with several telomere-associated proteins, including telomere repeat factors 1 (TRF1) and 2 (TRF2), subunits of the origin recognition complex (ORC), heterochromatin protein 1 (HP1), histone H3 trimethyl K9 (H3 K9me3), and members of the DNA-damage-sensing pathway. siRNA depletion of TERRA caused an increase in telomere dysfunction-induced foci, aberrations in metaphase telomeres, and a loss of histone H3 K9me3 and ORC at telomere repeat DNA. Previous studies found that TRF2 amino-terminal GAR domain recruited ORC to telomeres. We now show that TERRA RNA can interact directly with the TRF2 GAR and ORC1 to form a stable ternary complex. We conclude that TERRA facilitates TRF2 interaction with ORC and plays a central role in telomere structural maintenance and heterochromatin formation.”
TERRA plays a role in mammalian telomere length regulation
The 2010 publication Chromatin regulation and non-coding RNAs at mammalian telomeres reports “In eukaryotes, terminal chromosome repeats are bound by a specialized nucleoprotein complex that controls telomere length and protects chromosome ends from DNA repair and degradation. In mammals the “shelterin” complex mediates these central functions at telomeres. In the recent years it has become evident that also the heterochromatic structure of mammalian telomeres is implicated in telomere length regulation. — Impaired telomeric chromatin compaction results in a loss of telomere length control. Progressive telomere shortening affects chromatin compaction at telomeric and subtelomeric repeats and activates alternative telomere maintenance mechanisms. Dynamics of chromatin structure of telomeres during early mammalian development and nuclear reprogramming further indicates a central role of telomeric heterochromatin in organismal development. In addition, the recent discovery that telomeres are transcribed, giving rise to UUAGGG-repeat containing TelRNAs/TERRA, opens a new level of chromatin regulation at telomeres. Understanding the links between the epigenetic status of telomeres, TERRA/TelRNA and telomere homeostasis will open new avenues for our understanding of organismal development, cancer and ageing.”
TERRA regulates and can inhibit telomerase expression
The 2010 publication The non-coding RNA TERRA is a natural ligand and direct inhibitor of human telomerase reports “Telomeres, the physical ends of eukaryotes chromosomes are transcribed into telomeric repeat containing RNA (TERRA), a large non-coding RNA of unknown function, which forms an integral part of telomeric heterochromatin. — TERRA molecules resemble in sequence the telomeric DNA substrate as they contain 5′-UUAGGG-3′ repeats near their 3′-end which are complementary to the template sequence of telomerase RNA. Here we demonstrate that endogenous TERRA is bound to human telomerase in cell extracts. Using in vitro reconstituted telomerase and synthetic TERRA molecules we demonstrate that the 5′-UUAGGG-3′ repeats of TERRA base pair with the RNA template of the telomerase RNA moiety (TR). — In addition TERRA contacts the telomerase reverse transcriptase (TERT) protein subunit independently of hTR. In vitro studies further demonstrate that TERRA is not used as a telomerase substrate. Instead, TERRA acts as a potent competitive inhibitor for telomeric DNA in addition to exerting an uncompetitive mode of inhibition. Our data identify TERRA as a telomerase ligand and natural direct inhibitor of human telomerase. Telomerase regulation by the telomere substrate may be mediated via its transcription.”
In practical terms: There may be plenty of telomerase present on telomeres. That telomerase will do no good to lengthen the telomeres, however, as long as it is bound up by TERRA
    
Epigenetic factors are key in determining telomerase activation, e.g. transcription of the telomerase gene hTERT
As pointed out in the Barzilai video, genetics plays a role in telomere length determination. The centenarians tend to have certain mutations associated with the hTERT gene. It has also long been known that many environmental, lifestyle and personal historical factors are also strongly determinative of telomere lengths. See for example the blog entry Part2: lifestyle, dietary, and other factors associated with telomere shortening and lengthening. But the mechanisms through which such factors affect telomere lengths were not understood until recently. There is increasing evidence that the mechanisms affecting telomerase transcription are epigenetic and that TERRA is centrally involved.
The role of epigenetics in regulating telomeres has been suspected for some time and was laid out in the 2007 publication The epigenetic regulation of mammalian telomeres. Increasing evidence indicates that chromatin modifications are important regulators of mammalian telomeres. Telomeres provide well studied paradigms of heterochromatin formation in yeast and flies, and recent studies have shown that mammalian telomeres and subtelomeric regions are also enriched in epigenetic marks that are characteristic of heterochromatin. Furthermore, the abrogation of master epigenetic regulators, such as histone methyltransferases and DNA methyltransferases, correlates with loss of telomere-length control, and telomere shortening to a critical length affects the epigenetic status of telomeres and subtelomeres. These links between epigenetic status and telomere-length regulation provide important new avenues for understanding processes such as cancer development and ageing, which are characterized by telomere-length defects.”
“It is becoming increasingly apparent that epigenetic modifications including CpG methylation, histone methylation and acetylation is important for TERT transcriptional regulation(ref).” This in part has become clear through studies focusing on cancers. “The chromatin structure via the DNA methylation or modulation of nucleosome histones has recently been suggested to be important for regulation of the hTERT promoter. DNA unmethylation or histone methylation around the transcription start site of the hTERT promoter triggers the recruitment of histone acetyltransferase (HAT) activity, allowing hTERT transcription(ref). For example, for hepatocellular carcinoma (HCC) the fifth most common cancer worldwide, “Thus, we propose that hTERT is regulated by a combination of epigenetic mechanisms (DNA methylation and histone modifications) and by the transcription factor c-myc in HCC(ref).”
The 2011 publication Telomeres in cancer and ageing reports “Furthermore, telomere length and integrity are regulated by a number of epigenetic modifications, thus pointing to higher order control of telomere function. In this regard, we have recently discovered that telomeres are transcribed generating long, non-coding RNAs, which remain associated with the telomeric chromatin and are likely to have important roles in telomere regulation.” The authors are talking about TERRA. In the same publication “Telomeric chromatin is also enriched in epigenetic marks that are characteristic of constitutive heterochromatin, such as histone tri-methylation and DNA hypermethylation, which act as negative regulators of telomere length and telomere recombination [2].” Also, “We also demonstrated that the telomeres of iPS cells acquire the epigenetic marks of the telomeres of embryonic stem cells, among them a low density of trimethylated histones H3K9 and H4K20, and that in the iPS cells there is a loss of telomeric silencing and an increase in the abundance of TERRA (telomeric transcripts) levels [29].”
Normal cells and cancer cells have different epigenetic telomere maintenance mechanisms via silencing of TERRA or avoiding TERRA and using ALT
The 2008 publication Telomerase activity is associated with an increase in DNA methylation at the proximal subtelomere and a reduction in telomeric transcription reports: “Tumours and immortalized cells avoid telomere attrition by using either the ribonucleoprotein enzyme telomerase or a recombination-based alternative lengthening of telomeres (ALT) mechanism. Available evidence from mice suggests that the epigenetic state of the telomere may influence the mechanism of telomere maintenance, but this has not been directly tested in human cancer. Here we investigated cytosine methylation directly adjacent to the telomere as a marker of the telomere’s epigenetic state in a panel of human cell lines. We find that while ALT cells show highly heterogeneous patterns of subtelomeric methylation, subtelomeric regions in telomerase-positive cells invariably show denser methylation than normal cells, being almost completely methylated. When compared to matched normal and ALT cells, telomerase-positive cells also exhibit reduced levels of the telomeric repeat-containing-RNA (TERRA), whose transcription originates in the subtelomere. Our results are consistent with the notion that TERRA may inhibit telomerase: the heavy cytosine methylation we observe in telomerase-positive cells may reflect selection for TERRA silencing in order to facilitate telomerase activity at the telomere. These data suggest that the epigenetic differences between telomerase-positive and ALT cells may underlie the mechanism of telomere maintenance in human tumorigenesis and highlight the broad reaching consequences of epigenetic dysregulation in cancer.”
Grainyhead-like 2 is a telomerase gene transcription factor that epigenetically regulates cell lifespans
I continue to be amazed by the continuing discovery of new substances relevant to human longevity. In this case it is Grainyhead-like 2 (GRHL2). The December 2010 publication Grainyhead-like 2 enhances the human telomerase reverse transcriptase gene expression by inhibiting DNA methylation at the 5′-CpG island in normal human keratinocytes reports: “We recently identified Grainyhead-like 2 (GRHL2) as a novel transcription factor that binds to and regulates the activity of the human telomerase reverse transcriptase (hTERT) gene promoter. – In this study, we investigated the biological functions of GRHL2 and the molecular mechanism underlying hTERT gene regulation by GRHL2. Retroviral transduction of GRHL2 in normal human keratinocytes (NHK) led to a significant extension of replicative life span, whereas GRHL2 knockdown notably repressed telomerase activity and cell proliferation. — Using promoter magnetic precipitation coupled with Western blotting, we confirmed the binding of GRHL2 to the hTERT promoter and mapped the minimal binding region at -53 to -13 of the promoter. Furthermore, mutation analysis revealed the three nucleotides from -21 to -19 to be critical for GRHL2 binding. — Because hTERT expression is regulated in part by DNA methylation, we determined the effects of GRHL2 on the methylation status of the hTERT promoter. Senescent NHK exhibited hypermethylation of the CpG island, which occurred with the loss of hTERT expression. On the contrary, the promoter remained hypomethylated in GRHL2-transduced NHK, irrespective of cell proliferation status. — Also, knockdown of endogenous GRHL2 led to hypermethylation of the promoter. These results indicate that GRHL2 regulates the hTERT expression through an epigenetic mechanism and controls the cellular life span.”
The epigenetic reprogramming required to create induced pluripotent stem cells affects TERRA 
As usual there is more related research going on out there beyond what I can review here, including that reported in these selected publications:
Wrapping it up
Key take-away observations from this richness of materials are:
· Telomere lengths tend to get shorter with age but not necessarily uniformly either in time or within a population cohort. As Dr. Barzilai points out in the video, some centenarians have longer telomeres than other people 30 years their junior. Telomere lengths are regulated dynamically with complex epigenetic factors playing central roles.
· The epigenetic factors regulate telomere lengths via several pathways and a newly-discovered RNA called TERRA is a central actor in that regulation. One of the key mechanisms involved in GPC island methylation of the hTERT telomerase gene.
· This epigenetic regulation is how lifestyle and dietary factors affect telomere lengths, a matter I have discussed extensively in previous blog entries. Chronic stress leads to shorter telomeres; a stable and happy lifestyle leads to longer ones, etc.
· Since epigenetic factors affect telomere lengths and shortened telomere lengths themselves generate epigenetic changes that have multiple impacts on cell biology, it appears that both of the following statements are true “Aging drives telomere lengths” and “Telomere lengths drive aging.” It is a chicken-and-egg situation.
· This does not mean that interventions aimed at extending telomere lengths are necessarily anti-aging interventions. Such interventions can normalize lifespans of mice whose lives are shorter because of induced telomerase deficiency, but have not been shown to extend the lives of normal mice. See the blog entry Mouse age reversal – very interesting but misrepresented research.
· Personally, I think we are already past the time to give up the simplistic notion that normal people can take a telomerase-activator supplement and therefore assure having longer telomeres and live longer. Neither our basic understanding nor experimental studies bear that conjecture out. On the other hand there is much research pointing to simple and practical lifestyle and dietary interventions that are likely to keep telomeres longer. See my blog entry Part2: lifestyle, dietary, and other factors associated with telomere shortening and lengthening.
· We still have only an initial and incomplete understanding of how the epigenetic programming of telomeres related to TERRA works. That whole area of research is barely 3 years old though other aspects of telomere biology have been studied for decades. As we learn more about it, we may pick up additional practical tips on how to live longer. Avanti!
Posted in Uncategorized | 5 Comments

Alzheimer’s Disease Update – March 2011

This is another of a series of blog posts relating to Alzheimer’s Disease (AD). The earlier posts included New views of Alzheimer’s disease and new approaches to treating it, The social cost of Alzheimer’s disease and late-life dementia, Diet and cognition, Warding off Alzheimer’s Disease and things in my diet, and a short post Deconstructing Alzheimer’s Disease – role of mitochondria. This update picks up on some topics introduced in the earlier entries, particularly in the May 2010 blog entry Alzheimer’s Disease research update and in the July 2010 blog entry Alzheimer’s disease studies validate anti-aging firewalls suggestions. And I discuss a few key new topics as well.

A short primer on Beta-amyloid and Tau tangles

The major mechanisms of AD pathology that have been studied intensely over the recent years are the intercellular accumulation of beta-amyloid protein and the intra-cellular buildup of tau tangles. As background, I briefly characterize both of these phenomena which characterize AD.“Amyloid beta (Aβ or Abeta or beta amyloid) is a peptide of 36–43 amino acids that appears to be the main constituent of amyloid plaques in the brains of Alzheimer’s disease patients. Similar plaques appear in some variants of Lewy body dementia and in inclusion body myositis, a muscle disease. Aβ also forms aggregates coating cerebral blood vessels in cerebral amyloid angiopathy(ref).” Generally, the amount of amyloid plaques in the brain is used as a measurement of the severity of AD.

Processing of the amyloid precursor protein

Tau tangles [also known as Neurofibrillary tangles (NFTs)] are tangles of misfolded tau protein that occur in nerve cells in AD patients. Tau tangles are “aggregates of hyperphosphorylation tau that are most commonly known as a primary marker of Alzheimer’s Disease. Their presence is also found in numerous other diseases known as Tauopathies(ref).” Tau proteins play important roles in healthy nerve tissues. The normal function of tau is to support microtubules, physical scaffold structures within nerve cells. “Tau proteins are proteins that stabilize microtubules. They are abundant in neurons in the central nervous system and are less common elsewhere. When tau proteins are defective, and no longer stabilize microtubules properly, they can result in dementias, such as Alzheimer’s disease(ref).” The presence of amyloid beta is known to lead to tau tangles. “The pathologic hallmarks of Alzheimer’s disease (AD) include senile plaque, neurofibrillary tangles (NFTs), synaptic loss, and neurodegeneration. Senile plaque and NFTs are formed by accumulation of amyloid-β (Aβ) and hyperphosphorylated tau, respectively(ref).”

For a number of years much of not most research on AD therapies was focused on strategies for prevention or removal of Aβ from brain tissues. Later, prevention or removal of tau tangles became another focus of research. And more-recently there has been concern with how aberrant microglial activation might be an upstream cause of both Aβ plaques and tau tangles.

My coverage of topics here is necessarily selective. Other important research related to AD is discussed in earlier blog entries accessible via the above links.

Some properties of amyloid beta

– – Amyloid beta may not actually be responsible for the pathology of AD.  “The “amyloid hypothesis”, that the plaques are responsible for the pathology of Alzheimer’s disease, is accepted by the majority of researchers but is by no means conclusively established. Intra-cellular deposits of tau protein are also seen in the disease, and may also be implicated. The oligomers that form on the amyloid pathway, rather than the mature fibrils, may be the cytotoxic species.[6] – – An alternative hypothesis is that amyloid oligomers rather than plaques are responsible for the disease. Mice that are genetically engineered to express oligomers but not plaques (APPE693Q) develop the disease. Furthermore mice that are in addition engineered to convert oligomers into plaques (APPE693Q X PS1ΔE9), are no more impaired than the oligomer only mice.[7](ref)” “Moreover, the recent failure of Aβ lowering agents, such as tramiprosate (11) and flurbiprofen (2) in phase III clinical trials, suggests that there is a need to pursue other therapeutic approaches, including those that reduce the levels of pathological tau(ref).”

Plentiful amyloid beta plaques can be found in the brains of dead patients who showed no signs of AD or dementia. “– Most, if not all, people have amyloid plaques in the brain years before they develop clinical symptoms of Alzheimer’s. — “It’s not uncommon for us to determine that an older person is fully intact mentally only to find the presence of substantial Alzheimer’s pathology on examining that person’s brain after death,” says John C. Morris, M.D., the Harvey A. and Dorismae Friedman Distinguished Professor of Neurology and director of the ADRC and of the Harvey A. Friedman Center for Aging. “We suspect that Alzheimer lesions may be present in the brain long before we can detect any clinical symptoms.”(ref)” Again, the culprit may be certain oligomers, as indicated in certain of the citations listed below.

– – Amyloid beta plaques can show up in a brain in only a day with AD neuronal changes shortly following. As reported in a 2009 Science Daily article Alzheimer’s-Associated Plaques Can Form In A Day, And Alzheimer’s Symptoms Soon Follow “– The amyloid plaques found in the brains of Alzheimer’s disease patients may form much more rapidly than previously expected. Using an advanced microscopic imaging technique to examine brain tissue in mouse models of the devastating neurological disorder, researchers from the MassGeneral Institute for Neurodegenerative Disease (MGH-MIND), working with colleagues from Washington University School of Medicine, find that plaques can develop in as little as a day and that Alzheimer’s-associated neuronal changes appear soon afterwards. — Although plaques formed rarely, they could appear as little as 24 hours after a previous plaque-free image was taken. The new plaques were similar in appearance to those seen in the brains of Alzheimer’s patients and in the mouse models, and subsequent imaging showed little change in the size of plaques once they had formed. – — Examining neurons adjacent to plaques showed that the kind of changes associated with Alzheimer’s — distortions in the projections through which neuronal signals pass — appear rapidly and approach maximum effect within five days. — “These results confirm the suspicion we’ve had that plaques are a primary event in the glial and neuronal changes that underlie Alzheimer’s dementia.””

AD and the Sirtuin SIRT1

In the May 2010 blog post on AD, the first Section is entitled SIRT1 and Resveratrol and Alzheimer’s Disease. There, I cite preliminary evidence for a hypothesis that activation of the SIRT1 gene, such as possibly by resveratrol, could be a preventative or therapeutic strategy against AD. I wrote “I discussed how some researchers think activation of SIRT1 might confer a strong therapeutic effect for control of Alzheimer’s disease. Several review articles published in the last couple of years articulate that hypothesis and suggest a potential role for resveratrol in controlling AD. These articles include the March 2010 e-publication Resveratrol as a Therapeutic Agent for Neurodegenerative Diseases, the 2009 publication Resveratrol and neurodegenerative diseases: activation of SIRT1 as the potential pathway towards neuroprotection and the 2008 publication Modulation of sirtuins: new targets for antiageing.”

Since writing the previous blogs on AD, a new publication has appeared, SIRT1 Suppresses β-Amyloid Production by Activating the α-Secretase Gene ADAM10, one that reveals a mechanism through which SIRT1 activation combats AD. “A hallmark of Alzheimer’s disease (AD) is the accumulation of plaques of Aβ 1 40 and 1 42 peptides, which result from the sequential cleavage of APP by the β and -secretases. The production of Aβ peptides is avoided by alternate cleavage of APP by the α and -secretases. Here we show that production of β-amyloid and plaques in a mouse model of AD are reduced by overexpressing the NAD-dependent deacetylase SIRT1 in brain, and are increased by knocking out SIRT1 in brain. SIRT1 directly activates the transcription of the gene encoding the α-secretase, ADAM10. SIRT1 deacetylates and coactivates the retinoic acid receptor β, a known regulator of ADAM10 transcription. ADAM10 activation by SIRT1 also induces the Notch pathway, which is known to repair neuronal damage in the brain. Our findings indicate SIRT1 activation is a viable strategy to combat AD and perhaps other neurodegenerative diseases.”

The work characterized in this publication is discussed in a Science Daily article Gene Linked to Aging Also Linked to Alzheimer’s. “ScienceDaily (July 22, 2010) — MIT biologists report that they have discovered the first link between the amyloid plaques that form in the brains of Alzheimer’s patients and a gene previously implicated in the aging process, SIRT1. — The researchers found that SIRT1 appears to control production of the devastating protein fragments, termed A-beta peptides, that make up amyloid plaques.” [As described above by inducing cleavage of APP by the α and -secretases instead of by β and -secretases, yielding harmless protein fragments instead of Amyloid beta.] “They also showed that in mice engineered to develop Alzheimer’s plaques and symptoms, learning and memory deficits were improved when SIRT1 was overproduced in the brain, and exacerbated when SIRT1 was deleted.”

Potential control of AD via inhibition of 5-lipoxygenase

The November 2010 publication 5-lipoxygenase as an endogenous modulator of amyloid beta formation in vivo suggests a mechanism involved in the creation of beta amyloid plaques. “Objective : The 5-lipoxygenase (5-LO) enzymatic pathway is widely distributed within the central nervous system, and is upregulated in Alzheimer’s disease. However, the mechanism whereby it may influence the disease pathogenesis remains elusive. – Methods: We evaluated the molecular mechanism by which 5-LO regulates amyloid β (Aβ) formation in vitro and in vivo by pharmacological and genetic approaches.– Results: Here we show that 5-LO regulates the formation of Aβ by activating the cAMP-response element binding protein (CREB), which in turn increases transcription of the γ-secretase complex. Preventing CREB activation by pharmacologic inhibition or dominant negative mutants blocks the 5-LO-dependent elevation of Aβ formation and the increase of γ-secretase mRNA and protein levels. Moreover, 5-LO targeted gene disruption or its in vivo selective pharmacological inhibition results in a significant reduction of Aβ, CREB and γ-secretase levels. – Interpretation: These data establish a novel functional role for 5-LO in regulating endogenous formation of Aβ levels in the central nervous system. Thus, 5-LO pharmacological inhibition may be beneficial in the treatment and prevention of Alzheimer’s disease.”

Additional insight into the significance of this finding is contained in a 2010 Science Daily article Modulating a Protein in the Brain Could Help Control Alzheimer’s Disease. “According to Domenico Praticò, an associate professor of pharmacology in Temple’s School of Medicine and the study’s lead researcher, the 5-Lipoxygenase enzyme is found in abundance mainly in the region of the brain, the hippocampus, involved in memory. — Praticò and his team discovered that 5-lipoxygenase, which unlike most proteins in the brain increases its levels during the aging process. It also controls the activation state of another protein, called gamma secretase, a complex of four elements which are necessary and responsible for the final production of the amyloid beta, a peptide that when produced in excess deposits and forms plaques in the brain. — “What we found was 5-lipoxygenase regulates and controls the amount of total amyloid beta produced in the brain,” said Praticò. “With aging, the more 5-lipoxygenase you have the more amyloid beta you’re going to produce. This will translate into a higher risk to develop full Alzheimer’s.” — A previous study by Praticò, in which researchers crossed a mouse model of Alzheimer’s with a mouse that did not genetically feature 5-lipoxygenase, demonstrated that a lack of this enzyme protein alone can reduce the amount of disease in the brain by up to half. — “It has been known for years that the 5-lipoxygenase is an important protein in other areas of the body, such as the lung, but nobody really cared about its role in the brain,” he said. “Based on some previously know information, we questioned whether this enzyme was a primary or secondary player in the development of Alzheimer’s. What we found was a new primary role for an old enzyme.””

The significance of this finding is that drugs that inhibit 5-lipoxygenase are already on the market and could possibly be used to control AD. Continuing, “Praticò said that the key in the process was 5-lipoxygenase’s direct control over the gamma secretase, the only source of amyloid beta in the brain. “If you can modulate this enzyme easily, then you can control the amount of total amyloid beta that is produced by the gamma secretase in the brain, thus controlling the amount of Alzheimer’s disease.” — Praticò said that armed with new information, new therapies could be developed to block the increase of 5-lipoxygenase levels in the aging brain, which would in turn prevent the formation of amyloid beta. He said that there are several FDA-approved 5-lipoxygenase inhibitors currently being used for the treatment of asthma, and that the Temple researchers tested some of these inhibitors in the lab against the production of amyloid beta with initial positive results. — “These drugs are already on the market, they’re inexpensive and, most importantly, they are already FDA-approved, so you wouldn’t need to go through an intense drug discovery process,” said Praticò. “So you could quickly begin a clinical trial to determine if there is a new application for an old drug against a disease where there is currently nothing(ref).”

Microglial senescence and AD

In my February 2010 blog entry New views of Alzheimer’s disease and new approaches to treating it, I cited evidence for the hypothesis that microglial cell senescence is probably a fundamental cause for AD, a cause upstream of beta amyloid plaque production and the setting in of tau tangles. There seems to be accumulating evidence for this hypothesis.

First of all, activation of non-senescent microglia appears to play a positive role at least in the early stages of AD. The 2011 publication Determination of Spatial and Temporal Distribution of Microglia by 230nm-High-Resolution, High-Throughput Automated Analysis Reveals Different Amyloid Plaque Populations in an APP/PS1 Mouse Model of Alzheimer’s Disease reports “One early and prominent pathologic feature of Alzheimer’s disease (AD) is the appearance of activated microglia in the vicinity of developing β-amyloid deposits. However, the precise role of microglia during the course of AD is still under discussion. Microglia have been reported to degrade and clear β-amyloid, but they also can exert deleterious effects due to overwhelming inflammatory reactions. Here, we demonstrate the occurrence of developing plaque populations with distinct amounts of associated microglia using time-dependent analyses of plaque morphology and the spatial distribution of microglia in an APP/PS1 mouse model. In addition to a population of larger plaques (>700µm(2)) that are occupied by a moderate contingent of microglial cells across the course of aging, a second type of small β-amyloid deposits develops (≤400µm(2)) in which the plaque core is enveloped by a relatively large number of microglia. Our analyses indicate that microglia are strongly activated early in the emergence of senile plaques, but that activation is diminished in the later stages of plaque evolution (>150 days). These findings support the view that microglia serve to restrict the growth of senile plaques, and do so in a way that minimizes local inflammatory damage to other components of the brain.” The 2011 publication Mechanism mediating oligomeric Aβ clearance by naïve primary microglia provides insight into how microglia are involved in the clearance of Aβ and their roles in the early stages of AD.

Second, it appears that with aging microglia lose their capability to get rid of beta amyloid via phagocytosis. A March 2011 publication supports this case: Microglia Demonstrate Age-Dependent Interaction with Amyloid-β Fibrils. “Alzheimer’s disease (AD) is an age-associated disease characterized by increased accumulation of extracellular amyloid-β (Aβ) plaques within the brain. Histological examination has also revealed profound microglial activation in diseased brains often in association with these fibrillar peptide aggregates. The paradoxical presence of increased, reactive microglia yet accumulating extracellular debris suggests that these cells may be phagocytically compromised during disease. Prior work has demonstrated that primary microglia from adult mice are unable to phagocytose fibrillar Aβ1-42 in vitro when compared to microglia cultured from early postnatal animals. These data suggest that microglia undergo an age-associated decrease in microglial ability to interact with Aβ fibrils. In order to better define a temporal profile of microglia-Aβ interaction, acutely isolated, rather than cultured, microglia from 2 month, 6 month, and postnatal day 0 C57BL/6 mice were compared. Postnatal day 0 microglia demonstrated a CD47 dependent ability to phagocytose Aβ fibrils that was lost by 6 months. This corresponded with the ability of postnatal day 0 but not adult microglia to decrease Aβ immunoreactive plaque load from AD sections in vitro. In spite of limited Aβ uptake ability, adult microglia had functional phagocytic uptake of bacterial bioparticles and demonstrated the ability to adhere to both Aβ plaques and in vitro fibrillized Aβ. These data demonstrate a temporal profile of specifically Aβ-microglia interaction with a critical developmental period at 6 months in which cells remain able to interact with Aβ fibrils but lose their ability to phagocytose it.”

The 2011 publication γ-Secretase component presenilin is important for microglia β-amyloid clearance suggests another important role for γ-secretase beyond that already outlined above. “We suggest for the first time, a dual role for γ-secretase in Alzheimer’s disease. One role is the cleavage of the amyloid precursor protein for pathologic β-amyloid production and the other is to regulate microglia activity that is important for clearing neurotoxic β-amyloid deposits. Further studies of γ-secretase-mediated cellular pathways in microglia may provide useful insights into the development of Alzheimer’s disease and other neurodegenerative diseases, providing future avenues for therapeutic intervention.”

If someone were to tell me last week that smoking pot could help ward off Alzheimer’s Disease by affecting microglial activation, my response would likely have been “What have you been smoking lately?” That would have been my reaction before I found this February 2011 publication Cannabidiol and other cannabinoids reduce microglial activation in vitro and in vivo: relevance to Alzheimers’ disease. “Microglial activation is an invariant feature of Alzheimer’s disease (AD). Interestingly cannabinoids are neuroprotective by preventing β-amyloid (Aβ) induced microglial activation both in vitro and in vivo. On the other hand, the phytocannabinoid cannabidiol (CBD) has shown anti-inflammatory properties in different paradigms. In the present study we compared the effects of CBD with those of other cannabinoids on microglial cell functions in vitro and on learning behaviour and cytokine expression following Aβ intraventricular administration to mice, – Interestingly cannabinoids are neuroprotective by preventing β-amyloid (Aβ) induced microglial activation both in vitro and in vivo. On the other hand, the phytocannabinoid cannabidiol (CBD) has shown anti-inflammatory properties in different paradigms. In the present study we
compared the effects of CBD with those of other cannabinoids on microglial cell
functions in vitro and on learning behaviour and cytokine expression following
Aβ intraventricular administration to mice. CBD, WIN 55,212-2 (WIN), a mixed
CB(1)/CB(2) agonist, and JWH-133 (JWH), a CB(2) selective agonist,
concentration-dependently decreased ATP-induced (400 [micro]M) increase in
intracellular calcium ([Ca(2+)](i)) in cultured N13 microglial cells and in rat
primary microglia. In contrast HU-308 (HU), another CB(2) agonist, was without
effect. Cannabinoid and adenosine A(2A) receptors may be involved in the CBD
action. CBD and WIN-promoted primary microglia migration was blocked by CB(1)
and/or CB(2) antagonists. JWH and HU-induced migration was blocked by a CB(2)
antagonist only. All the cannabinoids decreased LPS-induced nitrite generation,
which was insensitive to cannabinoid antagonism. Finally both CBD and WIN,
following subchronic administration for three weeks.”

“Antioxidant activity has been reported to be a general property of the phenolic components of marijuana. Unlike Δ9-THC, cannabidiol can be administered at relatively high doses without undesired toxic or psychological effects.1(ref)” The March 2011 publication Caspase signalling controls microglia activation and neurotoxicity implicates caspases in microglial activation and therefore in AD. “Activation of microglia and inflammation-mediated neurotoxicity are suggested to play a decisive role in the pathogenesis of several neurodegenerative disorders. Activated microglia release pro-inflammatory factors that may be neurotoxic. Here we show that the orderly activation of caspase-8 and caspase-3/7, known executioners of apoptotic cell death, regulate microglia activation through a protein kinase C (PKC)-δ-dependent pathway. — We observe that these caspases are activated in microglia in the ventral mesencephalon of Parkinson’s disease (PD) and the frontal cortex of individuals with Alzheimer’s disease (AD). Taken together, we show that caspase-8 and caspase-3/7 are involved in regulating microglia activation. We conclude that inhibition of these caspases could be neuroprotective by targeting the microglia rather than the neurons themselves.”

To a considerable extent, aging-related neurodegenerative diseases appear to depend on the same underlying mechanisms

The previously-cited publication describes how caspase-induced microglial activation could be an underlying cause of both AD and Parkinson’s Disease. Another March 2011 publication brings multiple sclerosis under the same umbrella: Mechanisms of neurodegeneration shared between multiple sclerosis and Alzheimer’s disease. “Multiple sclerosis and Alzheimer’s disease are fundamentally different diseases. However, recent data suggest that certain mechanisms of neurodegeneration may be shared between the two diseases. Inflammation drives the disease in multiple sclerosis. It is also present in Alzheimer’s disease lesions, where it may have dual functions in amyloid clearance as well as in the propagation of neurodegeneration. In both diseases, degeneration of neurons, axons, and synapses occur on the background of profound mitochondrial injury. Reactive oxygen and nitric oxide intermediates are major candidates for the induction of mitochondrial injury. Radicals are produced through the induction of the respiratory burst in activated microglia, which are present in the lesions of both diseases. In addition, liberation of toxic iron from intracellular stores may augment radical formation. Finally reactive oxygen species are also produced in the course of mitochondrial injury itself. Anti-oxidant and mitochondria protective therapeutic strategies may be beneficial both in multiple sclerosis and Alzheimer’s disease in particular in early stages of the disease.”

The role of mitochondrial damage in AD is another topic of relevant in the chain of events leading to full-blown AD. I touched lightly on this in the early blog entry Deconstructing Alzheimer’s Disease – role of mitochondria. Another 2011 publication amplifies on the role of microglial activation in Parkinson’s Disease as well as in AD: CX3CL1 reduces neurotoxicity and microglial activation in a rat model of Parkinson’s disease. “The inflammatory response in the brain is tightly regulated at multiple levels. One form of immune regulation occurs via neurons. Fractalkine (CX3CL1), produced by neurons, suppresses the activation of microglia. CX3CL1 is constitutively expressed. — As hypothesized, CX3CL1 was able to suppress this microglia activation. The reduced microglia activation was found to be neuroprotective as the CX3CL1 treated rats had a smaller lesion volume in the striatum and importantly significantly fewer neurons were lost in the CX3CL1 treated rats. — These findings demonstrated that CX3CL1 plays a neuroprotective role in 6-OHDA-induced dopaminergic lesion and it might be an effective therapeutic target for many neurodegenerative diseases, including Parkinson disease and Alzheimer disease, where inflammation plays an important role.”

Other recent publications relating microglial activation to AD pathology
include:

* The increased density of p38 mitogen-activated protein kinase-immunoreactive microglia in the sensorimotor cortex of aged TgCRND8 mice is associated predominantly with smaller dense-core amyloid plaques (Feb 2011),

* Amyloid-β-induced reactive oxygen species production and priming are differentially regulated by ion channels in microglia (Feb 2011), and

* ATP and glutamate released via astroglial connexin 43 hemichannels mediate neuronal death through activation of pannexin 1 hemichannels (Feb 2011).

Exercise and tau AD pathology

The2011 publication Chronic exercise ameliorates the neuroinflammation in mice carrying NSE/htau23 concludes “In this study, the tau-transgenic (Tg) mouse, Tg-NSE/htau23, which over expresses human Tau23 in its brain, was subjected to chronic exercise for 3months, from 16months of age. The brains of Tg mice exhibited increased immunoreactivity and active morphological changes in GFAP (astrocyte marker) and MAC-1 (microglia marker) expression in an age-dependent manner. To identify the effects of chronic exercise on gliosis, the exercised Tg mice groups were treadmill run at a speed of 12m/min (intermediate exercise group) or 19m/min (high exercise group) for 1h/day and 5days/week during the 3month period. The neuroinflammatory response characterized by activated astroglia and microglia was significantly repressed in the exercised Tg mice in an exercise intensity-dependent manner. In parallel, chronic exercise in Tg mice reduced the increased expression of TNF-α, IL-6, IL-1β, COX-2, and iNOS. Consistently with these changes, the levels of phospho-p38 and phospho-ERK were markedly downregulated in the brain of Tg mice after exercise. In addition, nuclear NF-κB activity was profoundly reduced after chronic exercise in an exercise intensity-dependent manner. These findings suggest that chronic endurance exercise may alleviate neuroinflammation in the Tau pathology of Alzheimer’s disease.”

At least some symptoms of AD can be reversed

A November 2010 publication CBP gene transfer increases BDNF levels and ameliorates learning and memory deficits in a mouse model of Alzheimer’s disease reports “Here we show that amyloid-β (Aβ) accumulation, which plays a primary role in the cognitive deficits of AD, interferes with CREB activity. We further show that restoring CREB function via brain viral delivery of the CREB-binding protein (CBP) improves learning and memory deficits in an animal model of AD. Notably, such improvements occur without changes in Aβ and tau pathology, and instead are linked to an increased level of brain-derived neurotrophic factor. The resulting data suggest that Aβ-induced learning and memory deficits are mediated by alterations in CREB function, based on the finding that restoring CREB activity by directly modulating CBP levels in the brains of adult mice is sufficient to ameliorate learning and memory. Therefore, increasing CBP expression in adult brains may be a valid therapeutic approach not only for AD, but also for various brain disorders characterized by alterations in immediate early genes, further supporting the concept that viral vector delivery may be a viable therapeutic approach in neurodegenerative diseases.”

Below I discuss how curcumin could possibly contribute to normalization of CREB activity.

Inhibition or removal of tau tangles by manipulation of Hsp70 and Hsp27 chaperone proteins

The chaperone proteins Hsp70 and Hsp27 apparently can play roles in preventing or getting rid of tau tangles in brains affected by AD. This should not be surprising since tau tangles are the results of misfolded proteins and the key role of these chaperone proteins is to assure proper protein folding. As I pointed out in the blog entry HSP70 to the rescue, “ While you are at it, by the way, you might want to check out The Incorrect protein folding theory of aging discussed in my treatise. The basic notion is that stress often leads to the misfolding of proteins, a process that can accelerate with age creating dysfunctional conditions and vulnerability to a number of diseases.

Misfolded proteins cannot perform their intended functions and can create active mischief. In a nutshell, the role of the HSP70 heat shock proteins is to mobilize when large numbers of misfolded proteins show up due to stress, and to fold them up properly again. So, HSP70 proteins play important roles in health maintenance and possibly also in longevity.” As it turns out, manipulation of both Hsp70 and Hsp27 can indeed play a role in clearance of tau tangles, but how and when they can be manipulated to do this is fairly complicated.

The 2009 publication Chemical Manipulation of Hsp70 ATPase Activity Regulates Tau Stability relates “Alzheimer’s disease and other tauopathies have recently been clustered with a group of nervous system disorders termed protein misfolding diseases. The common element established between these disorders is their requirement for processing by the chaperone complex. It is now clear that the individual components of the chaperone system, such as Hsp70 and Hsp90, exist in an intricate signaling network that exerts pleiotropic effects on a host of substrates. Therefore, we have endeavored to identify new compounds that can specifically regulate individual components of the chaperone family. Here, we hypothesized that chemical manipulation of Hsp70 ATPase activity, a target that has not previously been pursued, could illuminate a new pathway toward chaperone-based therapies.– Using a newly developed high-throughput screening system, we identified inhibitors and activators of Hsp70 enzymatic activity. Inhibitors led to rapid proteasome-dependent tau degradation in a cell-based model. Conversely, Hsp70 activators preserved tau levels in the same system. Hsp70 inhibition did not result in general protein degradation, nor did it induce a heat shock response. We also found that inhibiting Hsp70 ATPase activity after increasing its expression levels facilitated tau degradation at lower doses, suggesting that we
can combine genetic and pharmacologic manipulation of Hsp70 to control the fate
of bound substrates. Disease relevance of this strategy was further established
when tau levels were rapidly and substantially reduced in brain tissue from tau
transgenic mice. These findings reveal an entirely novel path toward therapeutic
intervention of tauopathies by inhibition of the previously untargeted ATPase
activity of Hsp70.”

In other words the opposite of what was expected occurred. Instead of Hsp70 working to properly refold proteins in tau tangles, inhibition of Hsp70 worked to get rid of the tau tangles.

Clarification of this surprising result is provided in the Science Daily article Protein Inhibitor Helps Rid Brain Of Toxic Tau Protein. “The USF researchers originally thought activating Hsp70 would direct the chaperone protein to decrease the tau gone bad — preventing tau from stacking up into tangles inside cells involved in memory and destroying them. But instead of restoring tau to its normal supportive function, activating Hsp70 actually led to tau’s preservation and even more accumulation, Dickey said. “Basically we think the chaperone binds to the tau, and somehow in the process of trying to fix things decides to keep holding onto tau when it shouldn’t. So, activating Hsp70 is not necessarily what we want to do; we ultimately want to inhibit Hsp70 to promote the release or clearance of tau …to kill the bad tau.”

The December 2010 publication Phosphorylation Dynamics Regulate Hsp27-Mediated Rescue of Neuronal Plasticity Deficits in Tau Transgenic Mice relates “Molecular chaperones regulate the aggregation of a number of proteins that pathologically misfold and accumulate in neurodegenerative diseases. Identifying ways to manipulate these proteins in disease models is an area of intense investigation; however, the translation of these results to the mammalian brain has progressed more slowly. In this study, we investigated the ability of one of these chaperones, heat shock protein 27 (Hsp27), to modulate tau dynamics. Recombinant wild-type Hsp27 and a genetically altered version of Hsp27 that is perpetually pseudo-phosphorylated (3×S/D) were generated. Both Hsp27 variants interacted with tau, and atomic force microscopy and dynamic light scattering showed that both variants also prevented tau filament formation. However, extrinsic genetic delivery of these two Hsp27 variants to tau transgenic mice using adeno-associated viral particles showed that wild-type Hsp27 reduced neuronal tau levels, whereas 3×S/D Hsp27 was associated with increased tau levels. Moreover, rapid decay in hippocampal long-term potentiation (LTP) intrinsic to this tau transgenic model was rescued by wild-type Hsp27 overexpression but not by 3×S/D Hsp27. Because the 3×S/D Hsp27 mutant cannot cycle between phosphorylated and dephosphorylated states, we can conclude that Hsp27 must be functionally dynamic to facilitate tau clearance from the brain and rescue LTP; however, when this property is compromised, Hsp27 may actually facilitate accumulation of soluble tau intermediates.”

The December 2010 Science Daily article Dynamics of Chaperone Protein Critical in Rescuing Brains of Alzheimer’s Mice from Neuron Damage explains further “The researchers concluded that Hsp27 must be able to fluctuate between activated and de-activated states to succeed at clearing abnormal tau, thus preventing the protein from sticking together and building up excessively in the brain. In addition, Hsp27 can only be effective in helping maintain healthy tau turnover if the chaperone protein interacts with tau while it’s still soluble — before tau has developed into solid nerve-killing tangles. The chaperone protein cannot disrupt already formed tau tangles. — “In some circumstances, the activated chaperone protein may help stabilize and recycle tau, restoring the protein so it can do its normal job of supporting nerve cell structure,” Dr. Dickey said. “But when tau has become abnormally folded, activated Hsp27 may actually hold onto the bad tau without letting go, subverting tau’s release or clearance from the brain. In that case, it would be better to inhibit or deactivate Hsp27 to get rid of the tau.””

Clearance of tau protein via manipulation of chaperone proteins continues to be an active area of AD research. Again, I remain skeptical as to the prospects for such an approach to lead to a successful AD therapy.

Curcumin and AD

Relative to the last-mentioned citation, in the August 2010 blog entry Neurogenesis, curcumin and longevity I described how the dietary supplement curcumin can contribute to increased levels of brain-derived neurotrophic factor (BDNF) and normalizing CREB. “The 2005 publication Dietary curcumin counteracts the outcome of traumatic brain injury on oxidative stress, synaptic plasticity, and cognition relates to the effects of but not the process of neurogenesis in rat’s brains. “– Here, — we have examined the possibility that dietary curcumin may favor the injured brain by interacting with molecular mechanisms that maintain synaptic plasticity and cognition. The analysis was focused on the BDNF system based on its action on synaptic plasticity and cognition by modulating synapsin I and CREB. — Supplementation of curcumin in the diet dramatically reduced oxidative damage and normalized levels of BDNF, synapsin I, and CREB that had been altered after TBI. Furthermore, curcumin supplementation counteracted the cognitive impairment caused by TBI. These results are in agreement with previous evidence, showing that oxidative stress can affect the injured brain by acting through the BDNF system to affect synaptic plasticity and cognition.”

While this research was reported in the context of traumatic brain injury (TBI), to the extent that curcumin can restore levels of BDNF and CREB to normal, it is possible that its consumption could improve the symptoms of AD. There is a growing literature on the potential role of curcumin as an AD therapy. But how does it work? I mention here only the 2011 publication Curcuminoid Binds to Amyloid-β1-42 Oligomer and Fibril. “Studies of Alzheimer’s disease (AD) strongly support the hypothesis that amyloid-β (Aβ) deposition in the brain is the initiating event in the progression of AD. Aβ peptides easily form long insoluble amyloid fibrils, which accumulate in deposits known as senile plaques. On the other hand, recent work indicated that soluble Aβ oligomers, rather than monomers or insoluble Aβ fibrils, might be responsible for neuronal and synaptic dysfunction in AD. Curcumin, a low molecular weight yellow-orange pigment derived from the turmeric plant, has shown therapeutic effects in transgenic mouse models of AD. However, it remains unclear whether curcumin interacts directly with the Aβ oligomers. This study investigated any interaction between curcumin and Aβ oligomers such as globulomer and Aβ-derived diffusible ligand (ADDL). Globulomer was observed as a cluster of spherical structures by electron microscopic analysis, and ADDL was also detected as small spherical structures. Fluorescence analysis revealed a significant increase in the fluorescence of curcumin when reacted with both oligomers. Furthermore quartz crystal microbalance analysis showed significant frequency decreases in oligomer-immobilized electrodes following the addition of curcumin. These results strongly suggested that curcumin binds to Aβ oligomers and to Aβ fibrils. The association of curcumin with Aβ oligomers may contribute to the therapeutic effect on AD. Based on these findings, curcumin could provide the basis of a novel concept in AD therapies targeting Aβ oligomers.”

As is generally the case when I write about complex topics, my coverage of subtopics here is necessarily partial and focused. A search in the government database of research publications pubmed.org using the term “Alzheimer’s” reveals 30,922 publications.

Wrapping it up

The above publications together suggest a rich handful of observations including:

1. I wonder if trying to treat AD by prevention or removal of amyloid beta, or prevention or removal of tau tangles for that matter, is like trying to treat smallpox or measles as skin diseases. I believe there are upstream causes of AD having to do with aging and that both amyloid beta and tau tangles are consequential downstream phenomena. No wonder that the vast efforts to treat AD based on getting rid of Aβ or tau have so far come to naught.

2. Properties of microglia are definitely age and senescence-related lending credence to seeing microglial senescence as an underlying cause of AD.

3. Activation of aged microglia could underlie other age-related neurodegenerative diseases besides Alzheimer’s Disease including Parkinson’s Disease and multiple sclerosis.

4. Suggested in my treatise as theories of aging, Oxidative Damage and Mitochondrial Damage act as mechanisms in the aetiology of AD, probably as downstream consequences of more basic causes such as microglial senescence.

5. Regular exercise leads to decreased activation of microglia and astroglia in certain tau+ transgenic mouse strains, and decreased neuroinflammation. This suggests that regular exercise may be protective of those already experiencing tau pathology in AD.

6. Evidence continues to emerge supporting the power of curcumin for prevention or treatment of AD as well as a number of other neurological
pathologies.

7. In a series of blog entries including the recent one Aging and diseases, I have repeated an opinion that for diseases if aging including AD, there is unlikely to be any basic cure that does not address the processes of aging themselves. Microglial senescence is such a process. The increase of 5-lipoxygenase with age is another of many such processes affecting AD. What I have seen in the above-reviewed AD research has served to reinforce that
opinion.

MEDICAL DISCLAIMER

FROM TIME TO TIME, THIS BLOG DISCUSSES DISEASE PROCESSES. THE INTENTION OF THOSE DISCUSSIONS IS TO CONVEY CURRENT RESEARCH FINDINGS AND OPINIONS, NOT TO GIVE MEDICAL ADVICE. THE INFORMATION IN POSTS IN THIS BLOG IS NOT A SUBSTITUTE FOR A LICENSED PHYSICIAN’S MEDICAL ADVICE. IF ANY ADVICE, OPINIONS, OR INSTRUCTIONS HEREIN CONFLICT WITH THAT OF A TREATING LICENSED PHYSICIAN, DEFER TO THE OPINION OF THE PHYSICIAN. THIS INFORMATION IS INTENDED FOR PEOPLE IN GOOD HEALTH. IT IS THE READER’S RESPONSIBILITY TO KNOW HIS OR HER MEDICAL HISTORY AND ENSURE THAT ACTIONS OR SUPPLEMENTS HE OR SHE TAKES DO NOT CREATE AN ADVERSE REACTION.

Posted in Uncategorized | 6 Comments

Aging and diseases – video blog

Everyone knows that old age can lead to many diseases and problems.  And sooner or later one of those diseases or problems will kill everybody.    But, what exactly is the relationship of aging to diseases?  The question leads to surprisingly interesting answers.

First, please have a look at this video for opinions of some prominent scientists:

Here, I address the points in the video and expand on several key related points. 

Aging increases the probability of multiple diseases and conditions which enhance the probability of death, and the rate of increase accelerates with age. 

Nobody dies of old age per-se but most everybody will die from a disease or problem of old age.  In the video Dr Guarante mentioned several disease susceptibilities that increase radically with aging including the biggies of cancer, heart disease, dementia and diabetes.  And there are many other conditions of aging that also lead to disability and death. For example, loss of eyesight and hearing can lead to automobile accidents.  Age related neurological and muscular changes can affect balance and body control leading to falls that create serious damage, disabilities and more imminent death.  Among trauma patients “Increasing age was associated with higher mortality, an increased proportion of falls and fatal head or spine injuries(ref).”

Increasing lifespans implies increasing healthspans.

Animal experiments with multiple species ranging from roundworms to fruit flys to mice indicate that interventions that radically increase lifespans have a similar effect on healthspans.  See, for example, the blog entry New extraordinary longevity lessons from the nematode which chronicles how researchers over 20 years have managed to discover interventions that multiply both the lifespans and healthspans of nematode worms (C-elegans) by a factor of over seven. 

In humans the ratio of healthspans to lifespans has been historically increasing, not decreasing.     

We are not only evolving to live longer(ref)(ref) but also the proportion of our life spent in a healthy state is increasing.  This is the gist of the message in the 2004 publication CHANGES IN THE DISPARITIES IN CHRONIC DISEASE DURING THE COURSE OF THE TWENTIETH CENTURY by the Nobel-Prize winning economist Robert W. Fogel.  This fact is contrary to the fear that extending lives will lead to longer and longer periods of disability and skyrocketing medical costs. If this tendency were to continue long enough we could approach the state of the “One hoss shay” where most of us live full healthy active lives until one day we simply drop dead.  Personally, I intend to be one of those. 

Interventions that increase our lifespans might be the most direct approach to addressing most diseases of old age

The major diseases of aging occur in the biomolecular architecture or “remodeling” that occurs with advancing age in individuals on the organ, cellular, proteomic and epigenetic levels.  An age-related disease like Alzheimer’s Disease (AD) is not due to a bacterial bug that could be cured with any conceivable antibiotic.  Rather, the disease is associated with multiple and complex changes in gene expression, cells and tissues that go with aging.  So far, despite billions of dollars spent on research on AD, there is still nothing approaching being a cure.  See my May 2010 blog entry Alzheimer’s Disease research update.  (I am, by the way, currently working on an update reflecting the vast recent research efforts being devoted to AD.)

In animal models, an intervention that increases lifespan tends to postpone all of the disease susceptibilities and problems that occur with aging.  They still happen, but happen later.  This has led to a simple but powerful hypothesis:

The best way to get at most intractable diseases of aging is to go first after aging itself.    

Along with some highly respected researchers I very strongly suspect that if in fact a cure for AD is found, it will turn out also to be an anti-aging treatment.  Supportive of this point is the July 2010 publication SIRT1 Suppresses β-Amyloid Production by Activating the α-Secretase Gene ADAM10.  On the one hand “Our findings indicate SIRT1 activation is a viable strategy to combat AD and perhaps other neurodegenerative diseases.”  On the other hand SIRT1 activation is activation of the calorie restriction pathway known to be life-extending for a wide variety of species.  I will say more about this particular research when I write next about AD research.  This research, incidentally, originated in Dr Guarante’s lab at MIT, the Glenn Laboratory for the Science of Aging.

Finding a cure to a single disease of old age may or may not by itself have a big effect on average lifespans. 

If such a cure addresses epigenetic factors related to aging, such a cure might indeed increase average lifespans(ref).  And I believe that for aging-related diseases, any approach to a cure that does not address the epigenetic factors is doomed to fail.  That is why I am optimistic that a good chunk of the $31.2 billion currently being spent by the NIH on medical research may turn out to be research on aging – even if it is now labeled as cancer, dementia or other research(ref). 

Many of the current anti-aging interventions like use of Rapamycin were discovered accidentally, and I expect that pattern to continue.  Rapamycin was discovered from a random soil sample on Easter Island, an island known by locals as “Rapa Nui.”  That is how the drug got its name.  Later the mTOR gene was discovered because it was the gene that most reacted to rapamycin.  mTOR stands for “mammalian target of rapamycin.  And a little later yet it was discovered that inhibiting mTOR via rapamycin was life-extending across many species.  And now, rapamycin and the mTOR pathway are the subjects of intense research mainly addressed at curing diseases(ref).”   

I am willing to bet a healthy vegetarian dinner with anybody that if and as any disease treatment based on mTOR pathway inhibition emerges from the FDA approval gumball machine, that treatment will be life-extending.

 On the other hand, there are so many diseases and problems of old age that finding a cure for only one – say a specific cancer – is likely to affect average lifespan by only a few months unless that cure also addresses underlying processes of aging.  Replacing the fan belts in a 25 year-old car might keep the car running a while longer but all the other systems will be continuing to go downhill.

New anti-aging drugs will have to come on the market because they help treat or cure specific diseases.

This is because aging per-se is not regarded as a disease by the FDA and therefore drug approval cannot be based on anti-aging properties.  Besides, any clinical trial of a drug for anti-aging effects would have to go on 30 years or more and is therefore out of the question.  Our society and our institutions are simply not ready for us living much longer lives.  See the blog entry Getting the world ready for radical life extension.

Sirtris Pharmaceuticals, now a subsidiary of the drug company Glaxo Smith-Kline, may provide a model of how things will go for life-extending drugs.  “Sirtris is focused on discovering and developing proprietary, orally available, small molecule drugs with the potential to treat diseases associated with aging, such as Type 2 Diabetes. Our research focus is on modulating the sirtuins, a recently discovered class of enzymes involved in the aging process.”  The company currently has six clinical trials in its drug-development pipeline. 

I am willing to offer a second wager with anybody, again for a healthy vegetarian dinner.  My bet is that if and as any Sirtris disease treatment based on stimulating SIRT1 emerges from the FDA approval gumball machine, that treatment will be life-extending.

It is too bad that it is not possible now for anybody to develop, extensively test and market a drug simply because it is life-extending, but that is the name-of-the-game today.  That fact, however, may not turn out to be a major limitation because curing age-related diseases and life extension are so wrapped up with each other.

This video blog entry, like previous ones, is being brought to you in close collaboration with the filmmaker Robert Kane Papas.  I expect we will generate more of these blog entries structured around short video segments on aspects of longevity science.  Robert is the filmmaker who produced the recently-released film To Age or Not to Age. Robert captured hundreds of hours of interesting video in shooting the film over a 4-year period, including extensive interviews with a number of prominent aging-science researchers.  It was possible to incorporate only a small fraction of that interesting material in the film itself.  However, Robert will be identifying short but remarkable segments of materials both in the film and not in the film, and I will be remarking on them just as in this blog entry.  I expect the videos and the remarks will appear on both this site and on the film site To Age or Not to Age.

Posted in Uncategorized | 4 Comments

Radio interview on aging sciences

My colleague filmmaker Robert Kane Pappas and I spoke about the longevity sciences and their implications on The Power Hour Wednesday March 9.  The Power Hour with Joyce Riley is a syndicated radio program available nationally and internationally.   Advertising-free audio files for the conversation can be downloaded and heard by clicking

https://files.me.com/dotcalm9/75tthn.mp3  (Part 1)  https://files.me.com/dotcalm9/21d6o2.mp3 (Part2)

Robert was the scheduled guest.  However, after I called in about 15 minutes into the show, the hostess Joyce Riley invited me to stay on the line.  I did that participating with Robert in the discussion until the end of the show.  The Power Hour program is commercially-supported and our on-radio conversation was interspersed with advertising commercials including ones for health products.  I emphasize that I appeared as a call-in guest on the show, have no commercial links to or interests in any of the products advertised, and do not necessarily endorse their use.  I believe the same holds for Robert.  Also, to clear another matter up, Joyce referred to me as a “prominent geneticist,” which I am not.  I did point that out later in the show, indicating that my field is interpretation of advanced research in all fields of science relating to longevity.  Beyond that, Joyce asked a lot of provocative questions leading to a lively discussion. The Power Hour program is directed at a general audience of radio listeners, and Joyce repeatedly mentioned that there were large numbers of people calling in, “the switchboard is overloaded.”  Many of the call-in questions and remarks were interesting because they illustrate the lack of public information about aging.  Also evident in the caller remarks was the low esteem in which science is held by many people, assuming  for example that any products of scientific research are not “natural” and therefore against God’s will or the natural order of things.  Also evident was failure to distinguish between the activities of basic research scientists on the one hand and exploitative drug company practices on the other hand.  Nonetheless, some of the discussion relating to our health care system and the role of the pharmaceutical industry was interesting. 

Also interesting was the repeated raising of the question:  “If and as interventions for significantly extending human lifespans become available, will they be available to the general public or only to the very-rich?”

Robert has spent the last 4 years producing the film To Age or Not to Age and in the process has conducted extensive interviews as well as informal discussions with many prominent researchers in the longevity sciences.  And he and I have enjoyed many long and sometimes-heated discussions.  This has given Robert a unique perspective of the aging research area – that of a sensitive interviewer and a  filmmaker.

Shortly before the film To Age or Not to Age was due to be shown for the first time on national TV late last Fall, Robert came across this blog and decided he had somehow to shoehorn me into his film.   We met that weekend in Bridgeport Connecticut for an improvised filming session in a public park.  I appear in three short segments towards the end of the final film, briefly presenting my theory of how closing the loop in the stem cell supply chain could lead to very long lives.  Since then Robert and I have established a close collaborative relationship.  We find ourselves aligned on the need to better inform the public on a variety of issues connected with aging research and the personal and social implications of ever-longer lifespans.  Besides jointly bringing you the video entries in this blog and joining in events like this radio show, we have been planning other movies covering many aspects of the longevity sciences and the profound implications of us living longer lives.

A non-scientific but fun trailer for To Age or Not to Age is:

Posted in Uncategorized | 6 Comments

New name – same blog

As a belated second-birthday present, I am giving this blog a new name – Aging Sciences, as you can see in the header.   Starting now the latest blog entry will be available online at www.agingsciences.com.  Nothing is lost and all existing links to past articles will continue to work because they will retain their old addresses.  Users can continue to use the old blog address of www.anti-agingfirewalls.com if they prefer.   The old blog name basically references current anti-aging interventions, only one of many aspects of the aging puzzle.  The new name more-accurately reflects what the blog has grown up to be about – all the key sciences involved in the ongoing study of aging and possible interventions that can combat aging.  The new web address should also be a lot easier to remember.

Posted in Uncategorized | 3 Comments

The many faces of mTOR and rapamycin

The intent of this blog entry is to present a sampler of recent research relating to the mTOR pathway and the effects of rapamycin, focusing on recent and longevity-related results not covered in previous blog entries.
Background
Of course mTOR stands for mammalian target of rapamycin and the drug rapamycin inhibits the mTOR pathway. I have already written several blog entries related to the mTOR pathways. For a general introduction I suggest readers review Longevity genes, mTOR and lifespan. Other relevant past blog entries include Viva mTOR! Caveat mTOR! and More mTOR links to aging theories .
In my treatise one of the advanced “candidate” aging theories is Increasing mTOR signaling which happens with aging.” There, I described how the mTOR pathways appears to be highly conserved across species and how in primitive species as well as mice, inhibition of mTOR signaling is an effective strategy for extending longevity as well as addressing many disease processes.
The blog entry AMPK and longevity discusses the intimate relationship between the AMPK and mTOR pathways, and how activation of the former results in inhibition of the latter. The second part of the blog entry Curcumin, cancer and longevity contains a discussion of how curcumin inhibits mTOR expression and how feeding of rapamycin to mice is life-extending in mice. Other than for the material in this background section, my intent in this blog entry is to cover material not already covered.
I have previously written “Mammalian target of rapamycin (mTOR) is a protein encoded in humans by the FRAP1 gene. As the name suggests, mTOR is targeted by the immunosuppressive drug rapamycin, a drug used clinically to treat graft rejection and restenosis and being tested as a treatment for autoimmune diseases.   “The mTOR pathway integrates signals from nutrients, energy status and growth factors to regulate many processes, including autophagy, ribosome biogenesis and metabolism(ref, ref).”   The mTOR pathway is “a central controller of cellular and organism growth that integrates nutrient and hormonal signals, and regulates diverse cellular processes(ref).”
This diagram showing some of the key pathways related to mTOR appeared in the publication mTOR signaling: at the crossroads of plasticity, memory and disease:
As you may note, there are two related pathways involved commonly referred to as mTORC1 and mTORC2. mTORC1 is associated with Raptor. See The Mammalian Target of Rapamycin (mTOR) Partner, Raptor, Binds the mTOR Substrates p70 S6 Kinase and 4E-BP1 through Their TOR Signaling (TOS) Motif . Raptor (regulatory associated protein of mTOR) is a recently identified mTOR binding partner that also binds p70S6k and 4E-BP1 and is essential for TOR signaling in vivo” mTORC2 is in turn associated with Rictor, a different adaptor protein. Some inhibitors of mTOR affect only one of these pathways; others affect both. Likewise, disease processes may depend on interactions among the two pathways. For example, consider what is reported in the December 2010 publication Dual Inhibition of mTORC1/mTORC2 Induces Apoptosis of Mantle Cell Lymphoma by Preventing Rictor Mediated AKTS473 Phosphorylation by Potentiating AKT2-PHLPP1 Association. mTORC2 is largely resistant to rapamycin(ref).

A more complete layout of the biological roles of mTOR can be found in this diagram. And here
Clearly, the mTOR-related pathways are very complex and relate centrally to many critical biological functions.
mTOR and skeletal muscle
There is a body of literature going back some time concerned with the role of mTOR and other signaling pathways in the health and aging of skeletal muscle. See, for example, Skeletal muscle hypertrophy is regulated via AKT/mTOR pathway. “Skeletal muscle atrophies with disuse while with increased use and increased load skeletal muscle exhibits hypertrophy, with an increase in the size of existing muscle fibers. One signaling pathway involved in regulating skeletal muscle atrophy and hypertrophy is the AKT/mTOR pathway (see mTOR pathway). The mTOR pathway activity increases in response to muscle activity during hypertrophy and decreases in activity during atrophy. Blocking this pathway genetically or with the mTOR inhibitor rapamycin blocks hypertrophy and genetic activation of the pathway induces hypertrophy.”
mTOR and exercise
Consistent with the above, it appears that physical activity downregulates mTOR/S6K1 signaling and downregulates IRS-1 serine phosphorylation, at least in rat skeletal muscle.  See the 2010 publication Effects of Physical Actyivity and Nutritional Intake on Skeletal Muscle Protein Turnover and Cellular Signaling. So, could downregulation of mTOR be a pathway through which exercise increases longevity? It is an interesting conjecture.
Potential medical uses of rapamycin
Rapamycin is also known as sirolimus. It is marketed under the trade name Rapamune by Wyeth.” Rapamycin (sirolimus) is already in use as an immunosuppressant to prevent rejection of kidney transplants and as a treatment for psoriasis(ref).”
Possible new medical applications related to modulating the mTOR pathway
For several disease processes including sarcopenia, spinal cord injuries and epilepsy, researchers have been expressing hope that effective therapeutic interventions might be based on modulation of mTOR signaling.
mTOR and sarcopenia
Sarcopenia (from the Greek meaning “poverty of flesh”) is the degenerative loss of skeletal muscle mass and strength associated with aging (0.5-1% loss per year after the age of 25). Sarcopenia is a component of the frailty syndrome(ref).”
The 2011 publication Aging impairs contraction-induced human skeletal muscle signaling and protein synthesis reports “Sarcopenia, the loss of skeletal muscle mass during aging, increases the risk for falls and dependency. Resistance exercise (RE) training is an effective treatment to improve muscle mass and strength in older adults, but aging is associated with a smaller amount of training-induced hypertrophy. — This may be due in part to an inability to stimulate muscle-protein synthesis (MPS) after an acute bout of RE. We hypothesized that older adults would have impaired mammalian target of rapamycin complex (mTORC)1 signaling and MPS response compared with young adults after acute RE. — Conclusions: We conclude that aging impairs contraction-induced human skeletal muscle mTORC1 signaling and protein synthesis. These age-related differences may contribute to the blunted hypertrophic response seen after resistance-exercise training in older adults, and highlight the mTORC1 pathway as a key therapeutic target to prevent sarcopenia.”
The 2010 publication mTOR signaling as a target of amino acid treatment of the age-related sarcopenia reflects a hope and expectation that interventions in the mTOR pathway might be used to avert sarcopenia in elderly people. “Sarcopenia is an age-related structural and functional impairment of skeletal muscle leading to loss of strength, contractile capacity and endurance. Among factors implicated in sarcopenia, deregulation of muscle protein synthesis (MPS) has frequently been reported. Thus, the attempts aiming at identifying possible countermeasures to sarcopenia require consideration of a complex coordinated interaction of factors contributing to the balance between protein synthesis and breakdown and the identification of several regulators on their function. We will focus here on the signaling pathways controlling protein synthesis in skeletal muscle, specifically on one of the downstream effectors of the kinase Akt/PKB, the mammalian target of rapamycin (mTOR) kinase which is now recognized as a key regulator of cell growth and a pivotal sensor of nutritional status over the lifespan. Dysfunction of mTOR signaling in the elderly and its potential role as a target of amino acids in the treatment of age-related sarcopenia will be discussed.”
mTOR and spinal cord injury
The 2010 publication ATP-mediated protein kinase B Akt/mammalian target of rapamycin mTOR/p70 ribosomal S6 protein p70S6 kinase signaling pathway activation promotes improvement of locomotor function after spinal cord injury in rats suggests that interventions in the Akt/mTOR/p70S6K signaling pathway may improve recovery prospects after spinal cord injuries. “The protein kinase B (Akt)/mammalian target of rapamycin (mTOR)/p70 ribosomal S6 protein kinase (p70S6K) signaling pathway, as a central controller of cell growth, proliferation, survival, and differentiation in response to extracellular signals, growth factors, nutrient availability, energy status of the cell, and stress, has recently gained attention in neuroscience. The effects of this signaling pathway on repair of spinal cord injury (SCI), however, have not been well elucidated. ATP is increasingly recognized as an important regulator of signal transduction pathways, and plays important roles in functional recovery after nervous system injury. In the present study, we examined the ATP-induced changes of the Akt/mTOR/p70S6K signaling pathway in injured spinal cord of adult rats and potential therapeutic effects of this pathway on SCI-induced locomotor dysfunction. SCI was produced by extradural weight-drop using modified Allen’s stall with damage energy of 50 g-cm force. The rats were divided into four groups: SCI plus ATP, SCI plus saline, SCI plus ATP and rapamycin, and sham-operated. Using immunostaining studies, Western blot analyses and real-time qualitative RT-PCR analyses, we demonstrated that the Akt/mTOR/p70S6K signaling pathway is present in the injured spinal cord and the expression of its components at the protein and mRNA levels is significantly elevated by exogenous administration of ATP following SCI. We observed the effectiveness of the activated Akt/mTOR/p70S6K signaling pathway in improving locomotor recovery, significantly increasing the expression of nestin, neuronal nuclei (NeuN), neuron specific enolase (NSE), and neurofilament 200 (NF200), and relatively inhibiting excessive reactive astrogliosis after SCI in a rapamycin-sensitive manner.  We concluded that ATP injection produced a significant activation of the Akt/mTOR/p70S6K signaling pathway in the injured spinal cord and that enhancement of rapamycin-sensitive signaling produces beneficial effects on SCI-induced motor function defects and repair potential. We suggest that modulation of this protein kinase signaling pathway activity should be considered as a potential therapeutic strategy for SCI.”
mTOR and epilepsy
The 2009 publication Developing Antiepileptogenic Drugs for Acquired Epilepsy: Targeting the Mammalian Target of Rapamycin (mTOR) Pathway reports “A rational antiepileptogenic strategy is to target primary cell signaling pathways that initially trigger the downstream mechanisms causing epileptogenesis. Recent work implicates the mammalian target of rapamycin (mTOR) pathway as mediating epileptogenesis in a genetic epilepsy, Tuberous Sclerosis Complex (TSC), and suggests that mTOR inhibitors, such as rapamycin, may have antiepileptogenic properties for epilepsy in TSC. As mTOR regulates multiple cellular functions that may contribute to epileptogenesis in general, including ion channel expression, synaptic plasticity, and programmed cell death, mTOR inhibitors might also represent an effective antiepileptogenic therapy for other, more common types of epilepsy, such as acquired epilepsies due to brain injuries.”  The 2010 publication Mammalian target of rapamycin (mTOR) inhibition as a potential antiepileptogenic therapy: From tuberous sclerosis to common acquired epilepsies relates “The mammalian target of rapamycin (mTOR) pathway represents a logical candidate, because mTOR regulates multiple cellular functions that may contribute to epileptogenesis, including protein synthesis, cell growth and proliferation, and synaptic plasticity. The importance of the mTOR pathway in epileptogenesis is best illustrated by tuberous sclerosis complex (TSC), one of the most common genetic causes of epilepsy. In mouse models of TSC, mTOR inhibitors prevent the development of epilepsy and underlying brain abnormalities associated with epileptogenesis. Accumulating evidence suggests that mTOR also participates in epileptogenesis due to a variety of other causes, including focal cortical dysplasia and acquired brain injuries, such as in animal models following status epilepticus or traumatic brain injury. Therefore, mTOR inhibition may represent a potential antiepileptogenic therapy for diverse types of epilepsy, including both genetic and acquired epilepsies.”
An October 2010 report Pharmacological inhibition of the mammalian target of rapamycin pathway suppresses acquired epilepsy sounds a largely optimistic note: “Inhibition of mTOR by rapamycin has been shown to suppress seizures in TSC/PTEN genetic models. Rapamycin, when applied immediately before or after a neurological insult, also prevents the development of spontaneous recurrent seizures (epileptogenesis) in an acquired model. In the present study, we examined the mTOR pathway in rats that had already developed chronic spontaneous seizures in a pilocarpine model. We found that mTOR is aberrantly activated in brain tissues from rats with chronic seizures. Furthermore, inhibition of mTOR by rapamycin treatment significantly reduces seizure activity. Finally, mTOR inhibition also significantly suppresses mossy fiber sprouting. Our findings suggest the possibility for a much broader window for intervention for some acquired epilepsies by targeting the mTOR pathway.”
These reports are mostly based on mouse-model experiments involving a genetic TSC-related epilepsy. Another 2010 study report Regulation of cell death and epileptogenesis by the mammalian target of rapamycin (mTOR): A double-edged sword? sounds a note of caution. “Identification of cell signaling mechanisms mediating seizure-related neuronal death and epileptogenesis is important for developing more effective therapies for epilepsy. The mammalian target of rapamycin (mTOR) pathway has recently been implicated in regulating neuronal death and epileptogenesis in rodent models of epilepsy. In particular, kainate-induced status epilepticus causes abnormal activation of the mTOR pathway, and the mTOR inhibitor, rapamycin, can decrease the development of neuronal death and chronic seizures in the kainate model. Here, we discuss the significance of these findings and extend them further by identifying upstream signaling pathways through which kainate status epilepticus activates the mTOR pathway and by demonstrating limited situations where rapamycin may paradoxically increase mTOR activation and worsen neuronal death in the kainate model. Thus, the regulation of seizure-induced neuronal death and epileptogenesis by mTOR is complex and may have dual, opposing effects depending on the physiological and pathological context. Overall, these findings have important implications for designing potential neuroprotective and antiepileptogenic therapies that modulate the mTOR pathway.”
Finally, a 2011 study Rapamycin suppresses mossy fiber sprouting but not seizure frequency in a mouse model of temporal lobe epilepsy reports interesting additional insights “Temporal lobe epilepsy is prevalent and can be difficult to treat effectively. Granule cell axon (mossy fiber) sprouting is a common neuropathological finding in patients with mesial temporal lobe epilepsy, but its role in epileptogenesis is unclear and controversial. Focally infused or systemic rapamycin inhibits the mammalian target of rapamycin (mTOR) signaling pathway and suppresses mossy fiber sprouting in rats. We tested whether long-term systemic treatment with rapamycin, beginning 1 d after pilocarpine-induced status epilepticus in mice, would suppress mossy fiber sprouting and affect the development of spontaneous seizures. Mice that had experienced status epilepticus and were treated for 2 months with rapamycin displayed significantly less mossy fiber sprouting (42% of vehicle-treated animals), and the effect was dose dependent. However, behavioral and video/EEG monitoring revealed that rapamycin- and vehicle-treated mice displayed spontaneous seizures at similar frequencies. These findings suggest mossy fiber sprouting is neither pro- nor anti-convulsant; however, there are caveats. Rapamycin treatment also reduced epilepsy-related hypertrophy of the dentate gyrus but did not significantly affect granule cell proliferation, hilar neuron loss, or generation of ectopic granule cells. These findings are consistent with the hypotheses that hilar neuron loss and ectopic granule cells might contribute to temporal lobe epileptogenesis.”
Rapamycin and clinical trials
Although the above applications seem still-removed from clinical practice, a very large number of new specialized uses for rapamycin are in clinical trials. A search of clinicaltrials.gov using the term rapamycin reveals 639 clinical trials in various phases. Many of these relate to the immunosuppressant properties of rapamycin such as for prevention of transplant rejections and graft-host disease. A number relate to use of the substance on stents. Many relate to rapamycin as part of chemotherapy regimens for a large number of cancers. And some are investigating rapamycin for a wide variety of other diseases including Multiple Sclerosis, Polycystic Kidney Diseases, Tuberous Sclerosis; Lymphangioleiomyomatosis, In-Stent Restenosis, Oral Lichen Planus, Systemic Sclerosis, Angiomyolipoma, Diabetic Retinopathy, Aplastic Anemia, Glomerulosclerosis, Autoimmune Active Anterior Uveiti, and Age-Related Macular Degeneration. I conjecture that some or many of these trials will yield positive results and that therefore rapamycin will come into wider clinical use for a number of specialized applications.
Chronic use of rapamycin has not been seriously considered as an anti-aging treatment for humans because of safety concerns. One set of concerns has to do with the drug’s immunosuppressive properties. Another set of concerns has to do with its effects on metabolism.
Chronic rapamycin treatment can result in metabolic derangement

The 2010 publication Chronic Rapamycin Treatment Causes Glucose Intolerance and Hyperlipidemia by Upregulating Hepatic Gluconeogenesis and Impairing Lipid Deposition in Adipose Tissue reports “OBJECTIVE The mammalian target of rapamycin (mTOR)/p70 S6 kinase 1 (S6K1) pathway is a critical signaling component in the development of obesity-linked insulin resistance and operates a nutrient-sensing negative feedback loop toward the phosphatidylinositol 3-kinase (PI 3-kinase)/Akt pathway. Whereas acute treatment of insulin target cells with the mTOR complex 1 (mTORC1) inhibitor rapamycin prevents nutrient-induced insulin resistance, the chronic effect of rapamycin on insulin sensitivity and glucose metabolism in vivo remains elusive. — RESEARCH DESIGN AND METHODS To assess the metabolic effects of chronic inhibition of the mTORC1/S6K1 pathway, rats were treated with rapamycin (2 mg/kg/day) or vehicle for 15 days before metabolic phenotyping. — RESULTS Chronic rapamycin treatment reduced adiposity and fat cell number, which was associated with a coordinated downregulation of genes involved in both lipid uptake and output. Rapamycin treatment also promoted insulin resistance, severe glucose intolerance, and increased gluconeogenesis. The latter was associated with elevated expression of hepatic gluconeogenic master genes, PEPCK and G6Pase, and increased expression of the transcriptional coactivator peroxisome proliferator–activated receptor-γ coactivator-1α (PGC-1α) as well as enhanced nuclear recruitment of FoxO1, CRTC2, and CREB. These changes were observed despite normal activation of the insulin receptor substrate/PI 3-kinase/Akt axis in liver of rapamycin-treated rats, as expected from the blockade of the mTORC1/S6K1 negative feedback loop. — CONCLUSIONS These findings unravel a novel mechanism by which mTORC1/S6K1 controls gluconeogenesis through modulation of several key transcriptional factors. The robust induction of the gluconeogenic program in liver of rapamycin-treated rats underlies the development of severe glucose intolerance even in the face of preserved hepatic insulin signaling to Akt and despite a modest reduction in adiposity.”
Alternative inhibitors of rmTOR signaling

Besides rapamycin itself, a number of other substances to varying degrees also inhibit mTOR signaling. Some of the newer substances are reported in publications such as (2008) A new pharmacologic action of CCI-779 involves FKBP12-independent inhibition of mTOR kinase activity and profound repression of global protein synthesis, (2008) Palomid 529, a novel small-molecule drug, is a TORC1/TORC2 inhibitor that reduces tumor growth, tumor angiogenesis, and vascular permeability, (2009) Ku-0063794 is a specific inhibitor of the mammalian target of rapamycin (mTO and (2009) Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. Some of these alternative substances are selective in whether they affect mTORC1 or mTORC2. And they may vary also in toxicity and secondary effects.

The research literature relating to mTOR signaling is vast and rapidly growing. The topics covered in this blog entry only sample what is going on.

Posted in Uncategorized | 2 Comments