
Innovations in Science
614 grants match your search
-
nemaline myopathy - Alan Beggs, Ph.D.
Grant Announced: Winter 2016Disease Type: Endocrine Myopathies“Alan Beggs is working to uncover genes that cause nemaline myopathy.”
nemaline myopathy - Alan Beggs, Ph.D.
Alan Beggs, professor of pediatrics at Children’s Hospital Boston, was awarded an MDA research grant totaling $300,000 over a period of three years to uncover new genes that cause nemaline myopathy in patients and families where the underlying cause has not yet been identified. To better understand the biological pathways that lead to disease and to aid the search for effective therapies, Beggs will then develop zebrafish, mouse and dog models containing the newly uncovered mutations.
Funding for this MDA research grant began Feb. 1, 2016.
-
Congenital & Other Myopathies – Alan Beggs, PhD
Funded: 02/01/19 through 01/31/22Grant Announced: Winter 2019Disease Type: Congenital Myopathies“Alan Beggs, PhD, professor of Pediatrics at Harvard Medical School and director of the Manton Center for Orphan Disease ...”
Congenital & Other Myopathies – Alan Beggs, PhD
Alan Beggs, PhD, professor of Pediatrics at Harvard Medical School and director of the Manton Center for Orphan Disease Research at Children’s Hospital Boston, has been awarded an MDA research grant totaling $300,000 over three years to continue his previous research on the molecular genetics of congenital myopathies.
Congenital myopathies are a diverse group of inherited neuromuscular conditions caused by defects in skeletal muscle and include central core disease (CCD) and other core myopathies, nemaline myopathy, myotubular myopathy (MTM) and other centronuclear myopathies (CNMs), congenital myopathies with fiber-type disproportion (CFTDs), and others (some of these have not yet been identified). In previous MDA-funded work, Dr. Beggs built an extensive data registry and repository of samples from patients and their families. His screening for congenital myopathy-causing mutations in zebrafish led to the discovery of mutations in several genes that may be related to human neuromuscular diseases. His further analyses determined the nature of these mutations, their relationships to the muscle defects seen in zebrafish, and their relationships to the same genes in humans with analogous muscle findings.
In this new project, Dr. Beggs will use whole genome sequencing methods to discover the disease genes and genetic mutations that cause congenital myopathy in patients and families where the underlying cause has not yet been identified. Then, to better understand the biological pathways that lead to disease and search for effective therapies, Dr. Beggs will develop animal models with these mutations, which he will be able to use in validation studies (to test if the mutation actually causes the disease) and screening assays (to test small molecule libraries to identify new drugs to treat these conditions).
-
CMT — Bogdan Beirowski, M.D., Ph.D.
Grant Announced: Summer 2012Disease Type: Charcot-Marie-Tooth Disease (CMT)“Beirowski is studying the loss of axons in Charcot-Marie-Tooth disease (CMT). (Axons are the long fibers through which n...”
CMT — Bogdan Beirowski, M.D., Ph.D.
Postdoctoral research scholar Bogdan Beirowski, in the department of genetics at the Washington University School of Medicine in St. Louis, was awarded an MDA development grant totaling $180,000 over three years to study how defective Schwann cells lead to nerve-cell damage in Charcot-Marie-Tooth disease (CMT).
MDA development grants are awarded to exceptional postdoctoral candidates who have the best chance of becoming independent researchers and future leaders of neuromuscular disease research.
Degeneration of axons — fibers that carry electrical signals between the brain and spinal cord and the rest of the body — is a hallmark of CMT and some forms of Dejerine-Sottas disease, and is thought to be responsible for muscle weakness associated with these diseases.
In some forms of CMT, the primary molecular defect localizes to Schwann cells, which normally help nourish and protect axons by wrapping them in a multilayered membrane called myelin. But it's poorly understood how Schwann cell dysfunction causes axon damage.
Data from previous work suggests that removal of defective myelin (demyelination) causes inflammation that results in axon damage, but Beirowski hypothesizes that "defective Schwann cells deprive long axons of metabolic support, thus contributing to axon degeneration."
Beirowski has developed mouse models in which key metabolic regulators are blocked exclusively in Schwann cells, which he now will use to test whether failure of the cells to deliver nutrients to axons could explain axon loss.
The identification of specific metabolic defects in Schwann cells could lead to the development of CMT therapeutic strategies based on supporting the stability of axons.
Funding for this MDA grant began Aug. 1, 2012.
-
CMT – Bogdan Karl Beirowski, MD, PhD
Funded: 08/01/18 through 07/31/21Grant Announced: Summer 2018Disease Type: Charcot-Marie-Tooth Disease (CMT)“Axon degeneration is a central component of irreversible neurological disability in many neuromuscular diseases. There i...”
CMT – Bogdan Karl Beirowski, MD, PhD
“Axon degeneration is a central component of irreversible neurological disability in many neuromuscular diseases. There is accumulating evidence that enwrapping glia provide metabolic support to axons, and perturbed metabolic functions in these glia can result in axon degeneration.”
Bogdan Karl Beirowski, assistant professor at the University at Buffalo in Buffalo, New York, was awarded an MDA Research Grant totaling $300,000 over 3 years to study metabolic support of axons by LKB1 signaling in Schwann cells. Dr. Beirowski was the recipient of an MDA development grant in 2012.
Degeneration of long axons is a hallmark in neuropathies and neuromuscular diseases like Charcot-Marie-Tooth (CMT), Friedreich’s ataxia (FA), amyotrophic lateral sclerosis (ALS), and spinal muscular atrophy (SMA). Axonal losses lead to the debilitating neurological symptoms in these conditions, but underlying mechanisms are only poorly understood. Axons do not exist in isolation but are intimately associated with Schwann cells (SCs), which provide metabolic support to axons. When metabolic functions in these cells are disrupted, it can result in axon degeneration. In CMT neuropathies, it remains unknown how malfunction in SCs results in axon degeneration.
The Beirowski lab recently showed that SCs support integrity of long axons by virtue of their metabolism. Disruption of metabolic function in SCs from transgenic mice results in progressive axon degeneration, recapitulating a key component of human neuromuscular disease. Using genetic mouse models in his laboratory, together with the application of novel technologies to study SC metabolism, Dr. Beirowski and colleagues plan to identify glial metabolic pathways important for regulation of axon integrity and to investigate if abnormalities in such pathways may contribute to nerve damage and axon demise in CMT neuropathy models.
-
ALS – Michael Benatar, M.D., Ph.D., and Jonathan Katz, M.D.
Funded: 03/01/17 through 02/28/19Grant Announced: Additional Grants 2017Disease Type: Amyotrophic Lateral Sclerosis (ALS)““Integration of a mechanism to collect research-useful data through a clinical workflow within the electronic health r...”
ALS – Michael Benatar, M.D., Ph.D., and Jonathan Katz, M.D.
Michael Benatar, at the University of Miami Miller School of Medicine in Florida, and Jonathan Katz, at California Pacific Medical Center in San Francisco, were awarded a clinical research network grant to support their work on the Clinical Procedures To Support Research (CAPTURE) project, which aims to implement the “ALS Toolkit” within the Epic Electronic Health Record System. The ALS Toolkit provides a mechanism to systematize the collection of clinical data so that it can also be used for ALS (amyotrophic lateral sclerosis) research. The two-year award totaling $300,000 will support work conducted through Clinical Research in ALS and Related Disorders for Therapy Development (CReATe) aimed at lessening the burden on people with ALS to attend both clinical and research appointments. CReATe, a rare diseases clinical research consortium, is a member of the National Institutes of Health’s Rare Diseases Clinical Research Network.
Throughout the course of the disease, people with ALS require input and assistance from multiple health care professionals and, as a result, multidisciplinary clinics such as MDA ALS Care Centers are a key resource for ongoing medical management and care. Visits to care centers take several hours, and a substantial amount of clinical data (such as neurological examination results, quantification of respiratory muscle strength and motor function assessments) are routinely collected — much of it identical to the assessments performed at specialized research visits. However, data from these two types of visits are typically captured separately, necessitating separate appointments for people who, depending on the stage of their disease, may experience profound weakness, disability and difficulty associated with travel.
It has therefore been a longstanding goal of many ALS clinician-investigators to find ways to reduce the burden on patients by finding a way to regularly collect a standard data set at clinical appointments that can be used for research purposes as well.
With increasing penetration of the electronic medical record into academic medical centers, and the recently established collaboration between Benatar and Katz with Epic, a team will now develop and implement a novel ALS Toolkit (set of “smart forms”) within the electronic medical record system that will enable clinicians to collect standardized data that can serve both clinical and research purposes.
If successful, the work will support a fundamental change in practice that will provide people with ALS a simple and straightforward way to contribute to research that can accelerate progress towards treatments and cures.
-
ALS — Michael Benatar
Grant Announced: Summer 2010Disease Type: Amyotrophic Lateral Sclerosis (ALS)“"MDA funding has been absolutely critical for this study," Benatar said, noting that the "strength" of the study lies in...”
ALS — Michael Benatar
Michael Benatar, associate professor of neurology and epidemiology at Emory University in Atlanta, received an MDA grant totaling $525,000 to continue research into the early stage of FALS — familial, or inherited, ALS (amyotrophic lateral sclerosis, or Lou Gehrig's disease) — prior to symptom onset.
With funding from MDA, Benatar began his "pre-FALS" study in 2007, tracking a group of 30 people at risk for developing ALS because they harbor a mutation in the SOD1 gene, known to cause some forms of the disease. His team collected a series of physical, functional and neurological data over time in an effort to discern early biological markers ("biomarkers") of the disease process, and established a repository of biological samples collected from the study participants.
In Benatar's new follow-up study, the original group of 30 participants will expand to include presymptomatic individuals with mutations in other ALS susceptibility genes such as TDP43 and FUS.
Study aims include better definition of the presymptomatic stage of familial ALS, identification of environmental factors that might modify the age of disease onset among genetically susceptible individuals, continued development of biomarkers of early disease, and expansion of the existing collection of biological specimens.
Benatar's exploration into the presymptomatic phase of ALS could facilitate earlier recognition and diagnosis of both the familial and sporadic forms of the disease, which in turn could point the way toward therapeutics aimed at prevention or delay of ALS onset, as well as attempts to stop or slow the disease before it causes irreversible damage.
"MDA has shown great foresight in recognizing the importance of this study and the kind of long-term contribution that it can make to our understanding of the disease," Benatar said.
Funding for this MDA grant began August 1, 2010.
-
Clinical Procedures to Support Research (CAPTURE) - Michael Benatar, M.D., Ph.D.
Funded: 07/01/20 through 06/30/23Grant Announced: Summer 2020Disease Type: Amyotrophic Lateral Sclerosis (ALS)“A holy-grail of physician-investigators engaged in both ALS patient care and research has been the efficient and effecti...”
Clinical Procedures to Support Research (CAPTURE) - Michael Benatar, M.D., Ph.D.
"A holy-grail of physician-investigators engaged in both ALS patient care and research has been the efficient and effective use of clinically-derived data for research purposes. To address this critically unmet need, we have partnered with Epic (the maker of the most widely used electronic health record [EHR] system) to build the ALS Toolkit. This resource will permit broad sharing of de-identified clinical data in a regulatory-compliant manner."
Michael Benatar, Professor of Neurology at the Miller School of Medicine of the University of Miami, was awarded an MDA Clinical Research Network Grant totaling $916,261 over three years to support the multicenter deployment of the ALS toolkit, a module built into the Epic EHR that aims to facilitate structured data collection for research and improve quality of care.
Clinics are critical to ALS patient care, leading to improved quality of life and prolonged survival. Clinic visits can take several hours with substantial clinical data collected; however, these data have limited utility for research purposes because of the unstructured way in which the data are collected. The ALS toolkit will provide a platform for standardized data collection across clinical sites, which could obviate the need for separate research visits and reduce the burden on patients and physicians.
-
DM – Andrew Berglund, Ph.D.
Funded: 08/01/17 through 07/31/20Grant Announced: Summer 2017Disease Type: Myotonic Dystrophy (DM)“I think the myotonic dystrophy field has made great gains in understanding the mechanisms of this disease, and there a n...”
DM – Andrew Berglund, Ph.D.
“I think the myotonic dystrophy field has made great gains in understanding the mechanisms of this disease, and there a number of exciting therapeutic strategies underway,” Andrew Berglund says. “This includes clinical trials with antisense oligonucleotides that target the toxic RNA as well as other strategies that target other aspects of the disease.”
Andrew Berglund, professor of biochemistry and molecular biology at the University of Florida in Gainesville, received an MDA research grant totaling $300,000 over three years to develop a therapeutic strategy for both types 1 and 2 myotonic dystrophy (DM1, DM2).
It is thought that the genetic defect underlying DM causes a toxic RNA, which accumulates in the nucleus of the cell and causes protein dysfunction by trapping proteins important for normal cell function. (RNA is the chemical step between DNA and protein manufacturing.) Some therapeutic approaches aim at degrading these toxic RNAs, or prevent them from trapping important cellular proteins. Berglund’s approach is to prevent the toxic RNA from even being produced in the first place.
With colleagues, Berglund recently published proof-of-principle data supporting the approach they’ve developed, called IMPEDE (inhibition of microsatellite promoted expression of deleterious expansions), which showed that treatment with an FDA-approved drug called actinomycin D reduced toxic RNA production in DM1 cell and mouse models.
Now Berglund’s team is working to further develop this small-molecule approach. Small molecules have some advantages over other types of treatments — such as the ability to be given systemically and to get across the blood-brain-barrier.
If successful, this approach potentially could be used alone or in combination with other therapeutic strategies to treat myotonic dystrophy.
-
MMD/DM - Andrew Berglund, Ph.D.
Grant Announced: Spring 2014Disease Type: Myotonic Dystrophy (DM)“Andrew Berglund is investigating whether changing the shape of abnormal genetic material in type 1 or type 2 MMD can red...”
MMD/DM - Andrew Berglund, Ph.D.
Andrew Berglund, a professor of biochemistry, biophysics and molecular biology at the University of Oregon in Eugene, has been awarded an MDA research grant totaling $253,800 over three years to pursue changing the shape of the abnormal genetic material underlying type 1 and type 2 myotonic muscular dystrophy (MMD or DM). Berglund and colleagues will conduct laboratory experiments to see whether changing the shape of abnormally expanded genetic material (RNA) in these two forms of MMD can reduce or eliminate their harmful effects.
Funding for this MDA research grant began May 1, 2014.
-
DMD/BMD - Harold Bernstein, M.D., Ph.D.
Grant Announced: Winter 2011Disease Type: Duchenne Muscular Dystrophy (DMD)“(Left) A section of mouse leg muscle shows normal muscle fibers, with muscle cell nuclei shown in blue and aligned prope...”
DMD/BMD - Harold Bernstein, M.D., Ph.D.
Harold Bernstein, professor of pediatrics at the University of California, San Francisco, has been awarded an MDA research grant totaling $540,000 over three years. The award will help support Bernstein's study of human muscle development and potential cell-based therapies for treatment of degenerative muscle diseases such as Duchenne muscular dystrophy (DMD).
In his new work, Bernstein and colleagues will study the pathways that different types of muscle stem cells follow as they form new muscle, and identify the particular muscle stem cell types that appear most suited for therapeutic development.
The team will first observe the maturation of human stem cells into muscle cells in culture (in the laboratory), as a means of identifying the stages of normal muscle development. The investigators will then transplant the cells at various stages of development into the leg muscles of mice with a disease resembling DMD and study the process by which these cells become new muscle tissue, how this affects the animals' ability to exercise, and the strength of the treated muscles.
Bernstein's team hopes to fully elucidate the process of normal human muscle stem cell development and, in addition, identify specific stem cell types that may provide therapeutic benefit when transplanted into DMD-affected muscle.
"Funding from the Muscular Dystrophy Association will allow us to extend our research on muscle development and stem cell biology into studies specifically focused on therapy," Bernstein said. "This funding provides important support for research that will directly translate into improved treatment for patients with muscular dystrophy."
Funding for this MDA grant began February 1, 2011.
-
IBM - Sanford Bernstein, Ph.D.
Grant Announced: Winter 2012Disease Type: Inclusion-Body Myositis (IBM)“Pictured: Sanford Bernstein's lab group at San Diego State University. (Bernstein is standing in the back row, second fr...”
IBM - Sanford Bernstein, Ph.D.
Sanford Bernstein, a professor of biology at San Diego State University in California, was awarded an MDA research grant totaling $370,311 over a period of three years. The funds will help support Bernstein's research into the underlying molecular causes of, and potential treatments for, inclusion-body myopathy type 3 (IBM-3).
IBM-3 is caused by a mutation in a protein called myosin, the "molecular motor" that drives muscle contraction.
"Our biochemical and ultrastructural studies have shown that IBM-3 myosin is prone to unfolding and to forming clumps called aggregates," Bernstein said. "Further, muscle appears to respond to the mutant protein by producing autophagosomes, cellular bodies designed to encapsulate and degrade protein aggregates."
Bernstein's team will study the components that comprise the IBM-3 aggregates. Then, in an IBM-3 fruit fly research model that they developed, the investigators will test various approaches aimed at hastening the clearance of protein aggregates and ameliorating defective muscle structure and function in IBM-3.
"IBM-3 is a rare disease, and no model that produces the inclusion bodies we see in our drosophila model has been developed," Bernstein said. "Hence, use of this model presents the current best opportunity to develop a better understanding of the basis of the disease or its possible treatment."
Funding for this MDA grant began February 1, 2012.
-
ALS - James Berry, M.D.
Grant Announced: Winter 2011Disease Type: Amyotrophic Lateral Sclerosis (ALS)“Berry and colleagues are planning a phase 2 human clinical trial of the experimental drug ISIS-333611 as a follow-on stu...”
ALS - James Berry, M.D.
MDA has awarded a clinical research training grant totaling $180,000 to clinical research fellow James Berry at Massachusetts General Hospital (MGH) in Boston. The grant will support completion of a two-year fellowship during which Berry plans to study the effects of a drug called ISIS-333611 in familial, or inherited, ALS (amyotrophic lateral sclerosis, or Lou Gehrig’s disease).
Familial ALS accounts for 5 to 10 percent of all ALS cases. About 20 percent of those (1 to 2 percent of all ALS cases) are caused by a mutation in the SOD1 gene, which leads to production of abnormal SOD1 protein.
Berry is a member of the study team currently planning a phase 2 clinical trial of intrathecal (into the spinal canal) administration of the experimental drug ISIS-333611 (made by Isis Pharmaceuticals of Carlsbad, Calif.) in people with familial ALS. The trial will be a follow-on study to a phase 1 trial of the drug, currently under way, if results from that trial are favorable.
Previous studies of the drug in rats have shown that it is safe and tolerable, that it reduces mutant SOD1 RNA (the chemical step between DNA and protein synthesis) and protein levels, and slowed progression of the disease. It's thought that decreasing mutant SOD1 levels in humans may confer similar therapeutic benefits.
The planned phase 2 study will test ISIS-333611 over an extended period of time to assess its safety and tolerability. If the phase 2 trial is successful, a phase 3 trial may be planned to test efficacy of the drug.
"MDA funding is critical to my career development and my ability to begin my career as a clinical researcher in ALS," Berry said. "I am incredibly excited to have received this award, and I am looking forward to getting started on the project."
Funding for this MDA grant began February 1, 2011.
-
ALS - François Berthod, Ph.D.
Grant Announced: Winter 2011Disease Type: Amyotrophic Lateral Sclerosis (ALS)“This image shows mouse nervous system cells cultured in a three-dimensional environment. Motor neurons (red) and Schwann...”
ALS - François Berthod, Ph.D.
MDA has awarded a grant totaling $347,094 over three years to François Berthod, a professor in the department of surgery at Laval University in Quebec City, Quebec, Canada. The funds will help support Berthod's study of the underlying molecular mechanisms and disease process in ALS (amyotrophic lateral sclerosis, or Lou Gehrig's disease).
ALS is characterized by the degeneration of motor neurons (nerve cells that control muscle movement) in the spinal cord and brain.
Berthod, who has a background in tissue engineering, previously has shown that human motor neurons can differentiate (mature) from a population of stem cells obtained from human skin.
Now, Berthod plans to develop a three-dimensional model of the human spinal cord using neural (nervous system) cells obtained from the tissues of people with ALS. The human tissue-engineered spinal cord (TESC) model is expected to mimic the human disease "in vitro," or in a laboratory setting. Its design will permit testing of the interactions of various neural cell types, including motor neurons and neuroprotective cells such as astrocytes, microglia and Schwann cells, as a means of determining the conditions that induce or aid motor neuron death in ALS.
Findings from Berthod's work are expected to improve understanding of the causes and progression of sporadic ALS.
"MDA funding over the years has been the only source of support for our research on amyotrophic lateral sclerosis," Berthod said, adding that it's been "the strongest encouragement to continue developing innovative in vitro models of this disease."
Funding for this MDA grant began February 1, 2011.
-
DMD — Carmen Bertoni, Ph.D.
Grant Announced: Summer 2013Disease Type: Duchenne Muscular Dystrophy (DMD)“Carmen Bertoni, Ph.D., focuses on studying gene-editing strategies for DMD.”
DMD — Carmen Bertoni, Ph.D.
Carmen Bertoni, assistant professor of neurology at the University of California, Los Angeles, was awarded an MDA research grant totaling $300,000 over a period of three years to advance gene-editing strategies forDuchenne muscular dystrophy (DMD).
DMD is caused by a mutation in the dystrophin gene. Standard gene therapy techniques have focused on supplying an unmutated dystrophin gene to muscle cells. An alternative strategy, called gene editing, seeks to correct the mutant gene itself.
“Our research group [with MDA funding] has pioneered the use of gene editing strategies for the dystrophin gene to permanently correct the DNA,” Bertoni says.
The gene-editing toolkit is growing, and in this project, Bertoni will compare the correction efficiency of two different methods. Both specifically target the mutant sequence in the gene, and harness elements of the cell’s own “quality control” system to correct the mutation. The comparison will be performed first using muscle cells isolated from a mouse model for DMD, and then in the mice themselves, to determine the feasibility of using this technology in people.
The repair process is simple to describe, but complicated in practice. The therapy needs to reach many different muscle fibers, cross the dense and protective structure that surrounds every fiber, reach the cell nuclei and ultimately the dystrophin gene, and then direct the gene correction process. Finally, the amount of dystrophin expression achieved after correction needs to be sufficient to sustain muscle function over a prolonged period of time.
“Every one of these steps is necessary to ensure a safe and effective therapy in patients,” says Bertoni. “Altogether these studies have the potential to significantly advance the field of gene correction into a clinical scenario for the treatment of DMD.”
Funding for this MDA grant began August 1, 2013.
-
DMD/BMD — Carmen Bertoni
Grant Announced: Summer 2010Disease Type: Duchenne Muscular Dystrophy (DMD)“Under the microscope: Cross sections of muscle analyzed for dystrophin, the protein missing but needed in Duchenne muscu...”
DMD/BMD — Carmen Bertoni
MDA awarded $408,915 to Carmen Bertoni, assistant professor of neurology at the University of California, Los Angeles, to continue research into DNA repair strategies in Duchenne muscular dystrophy (DMD).
Bertoni's research group has pioneered a technology that uses small molecules called single-stranded oligonucleotides, or ssODNs, to act directly on the source of the problem in DMD: the "genetic blueprint," or DNA. In people with DMD, the DNA that makes up the dystrophin gene contains errors that lead to missing or nonfunctional dystrophin protein.
"We use ssODNs to let the muscle know of those errors and give the opportunity to each cell that composes muscles to correct the mistake," Bertoni said. The fix is permanent, negating the need for ongoing treatment.
The process involves injection of the oligonucleotide into muscle, where it seeks the flawed region of DNA and highlights the specific error. The cell is alerted to the presence of the genetic defect, and activates the repair process to correct the flaw.
To date, the main limitation of ssODN-mediated gene correction has been the low frequency of gene repair. Bertoni's group aims to increase the strategy's efficiency to generate levels of gene repair that are clinically meaningful as a viable treatment for DMD.
The group will test the oligonucleotides first in muscle cells and then, once investigators have optimized the structure, in dystrophin deficient mice.
"MDA has had and continues to have a pivotal role in the development of this technology for the treatment of DMD," Bertoni said. "Without any question, the advance of clinical applications to DMD using ssODNs heavily relies on the help that continues to be received by the MDA. It is a great honor to be part of the numerous and extremely talented scientists that are supported by the Association."
Funding for this MDA grant began August 1, 2010.
-
ALS, CMT - Martha Bhattacharya, Ph.D.
Grant Announced: Winter 2013Disease Type: Amyotrophic Lateral Sclerosis (ALS)“The peripheral nervous system of the fruit fly Drosophila melanogaster, shown with synaptic vesicle proteins in green an...”
ALS, CMT - Martha Bhattacharya, Ph.D.
Martha Bhattacharya, a postdoctoral research scholar in developmental biology at Washington University School of Medicine in St. Louis, Mo., was awarded an MDA development grant totaling $180,000 over a period of three years to study how and why axons degenerate.
Axons are the long extensions of motor neurons (muscle-controlling nerve cells) that link up with muscles. Signals are sent down the axon to cause the muscle to contract. When an axon degenerates, it can no longer carry those signals, leading to weakness.
“In neuromuscular diseases where motor neuron dysfunction is the primary cause of disability, such as amyotrophic lateral sclerosis (ALS) and Charcot-Marie-Tooth (CMT) disease, axonal degeneration is a unifying pathological hallmark of disease progression,” Bhattacharya says.
To study axonal degeneration, she and her colleagues developed a fruit fly research model that allows the identification of necessary components of the axonal degeneration cascade. Using this system, she has identified several key steps in the process, including one involving a protein called G-protein coupled receptor (GPCR), and another called protein kinase.
“These receptors are highly desirable drug targets,” Bhattacharya says, and pharmaceutical companies have a great deal of experience designing drugs to influence their behavior. “For the GPCR, we will determine its signaling mechanism in mammalian neurons and test its ability to protect neuromuscular synapses after injury. For the kinase, we will examine the effects of loss of this protein on mouse axons and synapses,” the sites of information exchange between nerve and muscle.
Learning more about the details of axonal degeneration also will help researchers understand more about the entire disease process, potentially leading to other targets for therapeutic intervention.
Funding for this MDA grant began Feb. 1, 2013.
-
FA - Sanjay Bidichandani, PhD
Funded: 02/01/19 through 01/31/22Grant Announced: Winter 2019Disease Type: Friedreich's Ataxia (FA)“The mechanism of gene silencing in FA is known as epigenetic silencing. As we dig deeper, we realize that the silencing ...”
FA - Sanjay Bidichandani, PhD
"The mechanism of gene silencing in FA is known as epigenetic silencing. As we dig deeper, we realize that the silencing mechanism is more complex than was initially appreciated. A fuller understanding of the epigenetic silencing mechanism will help yield therapeutic targets and perhaps even biomarkers to track therapeutic response in people with FA."
Sanjay Bidichandani, PhD, professor of Pediatrics at the University of Oklahoma Health Sciences Center, was awarded an MDA research grant totaling $300,000 over three years to study the role of epigenetic silencing of the frataxin gene (FXN) in Friedreich’s ataxia (FA).
In FA, mutations in the FXN gene cause decreased production of frataxin protein, resulting in diminished energy production in cells, including those of the nervous system and heart. MDA funding helped Dr. Bidichandani participate in research that led to the discovery of the FXN gene and its role in causing FA. In past work, Dr. Bidichandani discovered a DNA hypermethylation in FXN of people with FA. However, to identify effective therapeutic targets and biomarkers, a complete understanding of exactly how the silencing mechanism works is needed.
In this project, Dr. Bidichandani will look more closely at the two main mechanisms of FXN gene silencing to see if one mechanism precedes the other, or if they work in parallel. This information will inform if different therapeutic agents can work synergistically or limit the effectiveness of each other. Specifically, he will test the hypothesis that DNA hypermethylation acts as a barrier to efficient gene reactivation in FA, examining cells from people with FA and a humanized mouse model of the disease.
-
FA - Sanjay Bidichandani, M.B.B.S., Ph.D.
Grant Announced: Summer 2015Disease Type: Friedreich's Ataxia (FA)““We believe that this project will enhance our understanding of the molecular pathogenesis of Friedreich’s ataxia, a...”
FA - Sanjay Bidichandani, M.B.B.S., Ph.D.
CMRI Claire Gordon Duncan Chair in Genetics and Professor of Pediatrics Sanjay Bidichandani, at University of Oklahoma Health Sciences Center in Oklahoma City, was awarded an MDA research grant totaling $300,000 over three years to address clinically and scientifically important questions regarding the use of existing and novel HDAC inhibitors to increase levels of the frataxin protein in Friedreich’s ataxia (FA). Bidichandani and colleagues recently found that the mechanism of gene silencing in FA involves shutting off the FXN gene promoter, a region of the gene that typically controls when and how much a gene should be turned on. They are now working to identify therapeutic molecules that will effectively reverse FXN promoter silencing in FA, and to determine the precise mechanism by which promoter inactivation and reactivation occurs.
Funding for this MDA research grant began Aug. 1, 2015.
-
Jag1 gene therapy in muscular dystrophies – Stefano Biressi, PhD
Funded: 09/01/21 through 08/31/22Grant Announced: 2021Disease Type: Duchenne Muscular Dystrophy (DMD)“Despite the similar initiating cause, DMD patients vary significantly in the progression of the disease. Recent studies ...”
Jag1 gene therapy in muscular dystrophies – Stefano Biressi, PhD
"Despite the similar initiating cause, DMD patients vary significantly in the progression of the disease. Recent studies pointed at the membrane molecule jag1 as a positive modulator of muscle regeneration capable of conferring a nearly normal clinical phenotype and lifespan in naturally occurring jag1 overexpressing canine models of DMD."
Stefano Biressi, Ph.D., is an Associate Professor at the University of Trento in Provo-Trento, Italy, has been awarded an MDA Idea Grant totaling $25,000 for one year to increase jag1 membrane protein activity by using a novel class of synthetic long non-coding RNAs, also known as SINEUPs.
SINEUPs increase protein synthesis in mammalian cells by binding efficiently to target mRNA. They have been shown to upregulate protein synthesis within physiological levels, which shows an optimal safety profile. Dr. Biressi's work is to develop synthetic SINEUPs that can target the upregulation of jag1 in dystrophic mice. Their work will help speed up the process of developing a new approach to target other therapeutic genes in dystrophic muscle.
-
FSHD – Adam Bittel, PT, DPT, PhD
Funded: 06/01/19 through 05/31/21Grant Announced: Additional Grants 2019Disease Type: Facioscapulohumeral Muscular Dystrophy (FSH, FSHD)“We are interested in both understanding how exercise affects skeletal muscle in FSHD and identifying the key cellular ev...”
FSHD – Adam Bittel, PT, DPT, PhD
"We are interested in both understanding how exercise affects skeletal muscle in FSHD and identifying the key cellular events contributing to those responses."
Adam Bittel, PT, DPT, PhD, a postdoctoral fellow at Children’s National Medical Center in Washington, DC, was awarded the 2019 SSSI-MDA Fellowship Award. The award, co-sponsored by Strength, Science & Stories of Inspiration (SSSI) and MDA, will provide a total of $40,000 over two years to support Dr. Bittel’s work investigating the cellular mechanisms underlying the effects of exercise in facioscapulohumeral muscular dystrophy (FSHD).
During his doctoral studies, Dr. Bittel explored how exercise influences multi-organ metabolism in metabolic syndromes as well as in Barth syndrome, a genetic disease affecting skeletal and cardiac muscle. While exercise is known to improve muscle health and metabolism in other forms of muscular dystrophy, there are very few studies assessing the effects of exercise in individuals with FSHD. Dr. Bittel is interested in understanding how exercise affects skeletal muscle in a mouse model of FSHD (the FLExDUX4 mouse) and in identifying important cellular events contributing to those responses.
-
Glucocorticoid receptor and muscle stem cell quiescence – Alexandre Blais, PhD
Funded: 09/01/21 through 08/31/22Grant Announced: 2021Disease Type: Duchenne Muscular Dystrophy (DMD)“Although research has revealed a number of desirable properties for donor cells, in particular the ability to return to ...”
Glucocorticoid receptor and muscle stem cell quiescence – Alexandre Blais, PhD
"Although research has revealed a number of desirable properties for donor cells, in particular the ability to return to a state of dormancy and a location close to muscle cells, more work is needed to uncover methods to enforce the acquisition of these properties."
Alexandre Blais, Ph.D., is a Principal Investigator at the University of Ottawa in Ottawa, Ontario, Canada, has been awarded an MDA Idea Grant totaling $25,000 for one year to study the genome of muscle stem cells and identify roles of regulatory proteins.
Among the many potential therapies that are being investigated to treat neuromuscular diseases including Duchenne Muscular Dystrophy (DMD), muscle stem cell therapy provides promising approaches. However, there are still many challenges with muscle stem cell therapy that researchers are posed with. A common challenge is that donor cells do not assume the fate of desired cells once transplanted into recipient muscles. Dr. Blais’s research will help identify the molecular mode of action of a regulatory protein that is important for the dormancy of muscle stem cells.
-
DD/LGMD - Robert Bloch, Ph.D.
Grant Announced: Winter 2012Disease Type: Distal Myopathies“As part of their research, Bloch and colleagues will test shortened versions of the dysferlin protein for use in gene th...”
DD/LGMD - Robert Bloch, Ph.D.
MDA has awarded a research grant totaling $362,295 over a period of three years to Robert Bloch, a professor in the department of physiology at the University of Maryland School of Medicine in Baltimore. The funds will help support Bloch's study of the role of a protein called dysferlin in type 2B limb-girdle muscular dystrophy (LGMD2B) and distal muscular dystrophy (DD, or Miyoshi myopathy).
"Limb-girdle muscular dystrophy 2B and Miyoshi myopathy are caused by mutations in the gene that carries instructions for dysferlin, but the role of dysferlin and its location in muscle cells are controversial," Bloch said.
Bloch and colleagues are working to answer these questions by studying isolated muscle fibers from normal and dysferlin-mutant mice, studying the structure and function of intact muscles from these same mice, and comparing what they observe in mouse muscle to muscles of people with LGMD2B or Miyoshi myopathy.
"The state of research in my area is in considerable flux, largely because of problems that have arisen from attempts to translate studies done in isolated cells to what occurs in muscle tissue in a living animal," Bloch said. "We have been trying to bridge this gap and have been getting results that we did not expect based on earlier work from other laboratories.
"It will take us some time to sort out the differences, but once we do so we anticipate making rapid progress."
Funding for this MDA grant began February 1, 2012.
-
FSHD — Gregory Block, Ph.D.
Grant Announced: Summer 2013Disease Type: Facioscapulohumeral Muscular Dystrophy (FSH, FSHD)“Cells taken from an FSHD patient were coaxed to become early-stage muscle fibers called myotubes. The cells are labeled ...”
FSHD — Gregory Block, Ph.D.
Gregory Block, senior fellow in pediatrics at the University of Washington in Seattle, was awarded an MDA development grant totaling $179,994 over a period of three years to search for treatment targets infacioscapulohumeral muscular dystrophy (FSHD).
FSHD occurs when a section at the tip of one chromosome, chromosome number 4, is shortened. When that happens, it allows a gene called DUX4 to remain active into adulthood, instead of becoming inactive after early development. The symptoms of FSHD are due to this extended activity of DUX4, so preventing that activity, or blocking its effects, could be therapeutic in the disease. Block has developed a cell-based system to study DUX4 activation, and has found one chemical pathway in muscle cells that can prevent activation. He will continue investigating this pathway to determine if it can be used to develop treatments for FSHD.
“This is an exciting time for FSHD research,” Block says. “Four years ago, there was still considerable debate as to the genetic cause of the disease, and today we are challenged with defining pragmatic approaches for disease intervention. We are hoping the data generated from this project will provide considerable insight into the mechanisms used by cells to silence DUX4.”
Funding for this MDA grant began August 1, 2013.
-
Nemaline Myopathy – Jordan Blondelle, Ph.D.
Funded: 08/01/17 through 07/31/20Grant Announced: Summer 2017Disease Type: Endocrine Myopathies“My focus is to better understand the role that Cullin-3-related protein degradation may play in the pathological mechani...”
Nemaline Myopathy – Jordan Blondelle, Ph.D.
“My focus is to better understand the role that Cullin-3-related protein degradation may play in the pathological mechanism leading to the development of nemaline myopathy,” Jordan Blondelle says. “It’s important because the underlying mechanisms of this life-threatening disease currently are poorly understood.”
Jordan Blondelle, postdoctoral researcher at the University of California San Diego in La Jolla, was awarded an MDA development grant totaling $180,000 over three years to increase understanding of the underlying mechanisms in nemaline myopathy.
Muscle mass maintenance is tightly regulated by a balance between muscle growth and atrophy, reflecting the balance between protein synthesis and degradation at the cellular level. Accumulation of misfolded proteins or premature degradation of proteins is often associated with diseases such as skeletal muscle myopathies.
The degradation of muscle proteins is accomplished mainly through a cellular “clean-up and disposal system” called the ubiquitin-proteasome system, or UPS. The Cullin family of proteins, a key component of the UPS system, incorporate into complexes after which they are able to bind and mark unwanted proteins for degradation. Recently, mutations in several binding partners of Cullin-3, a Cullin protein family member, were found in people with severe forms of nemaline myopathy. These findings suggest a specific and important function for Cullin-3 targeted protein degradation during muscle development and maintenance.
Using mutant mice in which Cullin-3 is depleted in muscles, Blondelle and colleagues determined that the protein is necessary for survival, and will now look at how the loss of Cullin-3 affects muscle development, structure and function.
The team expects to find new target proteins of the Cullin-3 complex that would be misregulated in the absence of Cullin-3 and responsible for the myopathy observed in mice. In addition, the team intends to decipher, for the first time, how protein degradation mediated by Cullin-3 is relevant for muscle physiology, and to provide insight into the pathological mechanisms leading to nemaline myopathy in people.
If successful, Blondelle’s work could inform the development of therapies for nemaline myopathy.
-
CMT – Alessandra Bolino, PhD
Funded: 08/01/18 through 07/31/21Grant Announced: Summer 2018Disease Type: Charcot-Marie-Tooth Disease (CMT)“Our research is aimed at unravelling how myelination is regulated in the PNS and how perturbation of these mechanisms ca...”
CMT – Alessandra Bolino, PhD
“Our research is aimed at unravelling how myelination is regulated in the PNS and how perturbation of these mechanisms can cause disease in humans.”
Alessandra Bolino, head of the Human Inherited Neuropathies Unit at IRCCS Ospedale San Raffaele in Milano, Italy, was awarded an MDA Research Grant totaling $300,000 over 3 years to study the targeting of phospholipid and cytoskeleton dynamics to ameliorate CMT neuropathies.
Charcot-Marie-Tooth type 4B1 is a very severe demyelinating neuropathy with childhood onset, characterized by myelin outfoldings, redundant loops of myelin in the nerve that degenerate causing axonal problems. Dr. Bolino’s research previously demonstrated that loss of MTMR2 (Myotubularin-related 2) phosphatase is the cause of the disease. MTMR2 acts on phospholipids, important regulators of membrane trafficking, which is a key process in Schwann cells, the glial cells forming myelin in the peripheral nervous system. Dr. Bolino and colleagues also generated in vitro and in vivo models of CMT4B1, which have been instrumental over the years to study the pathophysiology of this neuropathy.
By investigating the mechanism by which loss of MTMR2 leads to altered membrane trafficking and myelin outfoldings in Schwann cells, they identified a novel signaling pathway that regulates the cytoskeleton dynamics and membrane growth within myelin-forming cells. In this project, they will further characterize this novel pathway and test whether this pathway is a good drug target. These approaches can be extended to other forms of CMT that are characterized by myelin outfoldings (CMT4B2, B3, CMT4C, CMT4H).
-
DMD - Justin Boyer, PhD
Funded: 02/01/19 through 01/31/22Grant Announced: Winter 2019Disease Type: Duchenne Muscular Dystrophy (DMD)“Our research will help define the role of satellite cells in muscular dystrophy. The findings from our proposed work wil...”
DMD - Justin Boyer, PhD
"Our research will help define the role of satellite cells in muscular dystrophy. The findings from our proposed work will have implications on therapeutics that are currently being tested for Duchenne muscular dystrophy as well as offer new a therapeutic strategy to treat other types of muscular dystrophies."
Justin Boyer, PhD, research fellow at the Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, was awarded an MDA development grant totaling $210,000 over three years to study the role of ERK1/2 signaling in satellite cells of mice models of Duchenne muscular dystrophy (DMD).
Extracellular signal-regulated kinases (ERKs) are proteins that are intricately involved in cell division. Normally, skeletal muscle can repair itself after injury with the help of a specific type of muscle stem cell called a satellite cell (SC). In previous work, Dr. Boyer found that the ERK1/2 protein signaling pathway is an essential mediator of SC survival and function. Silencing this pathway in SCs led to the loss of these cells in skeletal muscle, preventing it from recovering from injury. However, activating the ERK1/2 signaling pathway led to a greater number of SCs.
In this research, Dr. Boyer will further characterize ERK signaling, determine if increasing ERK1/2 activity increases satellite cell numbers, and determine if increased ERK1/2 activity improves muscle regeneration. In doing so, he hopes to further define the role of SCs in muscular dystrophy to better understand how these cells can be targeted for developing therapies to treat this disease.
-
DMD/BMD - Andrew Brack, Ph.D.
Grant Announced: Winter 2011Disease Type: Duchenne Muscular Dystrophy (DMD)“Brack studies adult muscle stem cells called "satellite cells," and seeks to determine what role, if any, this type of c...”
DMD/BMD - Andrew Brack, Ph.D.
Andrew Brack, an assistant professor at the Center for Regenerative Medicine at Massachusetts General Hospital in Boston, was awarded an MDA research grant totaling $353,259 over three years. The funds will help support Brack's research into the function of adult muscle stem cells called "satellite cells" in Duchenne muscular dystrophy (DMD) and possibly in other muscular dystrophies as well.
Satellite cells are responsible for tissue maintenance and repair over a lifetime. It's widely accepted among scientists that these cells are sufficient to promote muscle repair; however, it is unknown what is required of them for regeneration of diseased skeletal muscle and whether the number and function of cells in the satellite cell pool can be manipulated as a means of slowing or ameliorating disease progression.
Brack and colleagues plan to use targeted genetic approaches to manipulate the number and function of adult satellite cells in a research mouse model of DMD. From this they hope to gain an understanding of the relative importance of both number and functionality of satellite cells needed for effective muscle repair.
Results from Brack's work may shed light on new therapeutic strategies for slowing disease progression that are based on retaining a functional pool of satellite cells in people with DMD and other muscular dystrophies.
"Without funding through MDA," Brack said, "this project would not have been realized."
Funding for this MDA grant began February 1, 2011.
-
CMS — Paul Brehm, Ph.D.
Grant Announced: Summer 2012Disease Type: Congenital Myasthenic Syndromes (CMS)“Brehm and Dr. Mike Linhoff at their array tomography setup, where they are generating high-resolution images of the zebr...”
CMS — Paul Brehm, Ph.D.
Paul Brehm, senior scientist at Oregon Health Science University in Portland, was awarded an MDA research grant totaling $351,648 over three years to study the underlying mechanisms of movement disorders in some forms of congenital myasthenic syndromes (CMS).
Brehm’s research team has identified zebrafish research models that carry mutations in the same proteins that are known to cause some forms of CMS in humans. In both fish and humans, these proteins are localized to the neuromuscular junction (the space where signals are transferred between nerve and muscle). Their deficiency results in movement disorders such as use-dependent fatigue (the inability to perform repetitive tasks).
In one model, known as the rapsyn-deficient line (in which the fish have mutations in the gene for the rapsyn protein), Brehm and his team have been exploring the basis of use-dependent fatigue.
Rapsyn deficiency has been associated with problems on the muscle side of the neuromuscular junction. Brehm’s team has found that the absence of an as-yet unidentified signal produced by muscle regulates the motor neuron and that, ultimately, the absence of this signal appears to be accountable for use-dependent fatigue.
Brehm’s team is working to uncover the mechanism underlying the reduced motor neuron activity that precedes use-dependent fatigue, as well as ways in which to rescue the nerve defect.
“The means to study neuromuscular function using the zebrafish system has been unparalleled among the vertebrates,” Brehm says. “The discovery of zebrafish models that carry these mutant proteins now opens the way to addressing directly the consequences in humans.”
Funding for this MDA grant began Aug. 1, 2012.
-
Myofibrillar Myopathy – Robert Bryson-Richardson, Ph.D.
Funded: 02/01/18 through 01/31/20Grant Announced: Winter 2018Disease Type: Myofibrillar Myopathies (MFM)“We are testing multiple drugs in our animal model,” Robert Bryson-Richardson said. “Some of these, if effective, cou...”
Myofibrillar Myopathy – Robert Bryson-Richardson, Ph.D.
“We are testing multiple drugs in our animal model,” Robert Bryson-Richardson said. “Some of these, if effective, could be suitable for clinical use. Those that are not suitable for clinical use will help us to identify pathways that could be targeted in future therapies.”
Robert Bryson-Richardson, associate professor at the School of Biological Sciences, Monash University, in Victoria, Australia, was awarded an MDA research grant totaling $273,962 over three years to investigate potential approaches to treating myofibrillar myopathy (MFM) caused by a mutation in the Filamin C gene.
With colleagues, Bryson-Richardson has generated zebrafish models for MFM caused by a Filamin C mutation that reproduce key symptoms of the disease including accumulation of Filamin C protein into aggregates (clumps) and loss of muscle integrity, leading to muscle weakness. Using the models, the team is investigating two potential approaches to treat the disease: increasing the level of functional Filamin C protein overall so that there is more present in the muscle to compensate for the loss of some of it to aggregates, and removing the protein aggregates to slow down the sequestration of Filamin C.
In addition, the team is using CRISPR/Cas9 gene editing technology to develop new zebrafish models for the disease and screen for drugs that potentially could provide clinical benefit by increasing the level of functional protein or removing protein aggregates.
-
ALS - Antonius Bunt
Funded: 01/01/17 through 07/01/18Grant Announced: Additional Grants 2017Disease Type: Amyotrophic Lateral Sclerosis (ALS)“"This MDA grant is important for moving our IZ10023 platform technology forward, with potential benefits for not only pe...”
ALS - Antonius Bunt
Izumi Biosciences in Lexington, Mass., was awarded an MDA Venture Philanthropy (MVP) grant totaling $96,360 to fund early-stage development of a type of IZ10023, a type of drug called a “pharmacokinetic (PK) enhancer,” for use in people with ALS (amyotrophic lateral sclerosis) who are taking riluzole. (Riluzole, marketed under the brand name Rilutek, was developed based on MDA-supported research on the neurotransmitter glutamate, and is the only drug approved by the U.S. Food and Drug Administration to treat ALS.)
IZ10023 targets and blocks two proteins in the brain and spinal cord that work to protect the central nervous system (CNS) by acting as pumps that remove foreign or toxic substances. In mouse studies it’s been shown that these pumps remove drugs such as riluzole, reducing levels of the drug in the CNS and thereby limiting its effectiveness.
President and CEO of Izumi Biosciences Antonius Bunt will serve as the principal investigator on the project.
“Blockade of these pumps maintains effective levels of riluzole in the diseased tissues and leads to improved outcomes in animal models of ALS and other brain diseases where multi-drug resistance is a key clinical bottleneck,” Bunt explained.
Confirmation that the pumps’ activity is increased in most ALS patients and demonstration that sufficient blood levels of IZ10023 can be achieved to block their activity are important milestones that must be achieved before moving IZ10023 into clinical trials in patients with ALS, Bunt noted. If the work is successful, Izumi Biosciences intends to open an Investigational New Drug Application and move into clinical trials — possibly as early as 2019.
-
CMT - Robert Burgess, Ph.D.
Grant Announced: Summer 2015Disease Type: Charcot-Marie-Tooth Disease (CMT)“Robert Burgess is interested in developing mouse models of CMT and related neuromuscular disorders, and using these mice...”
CMT - Robert Burgess, Ph.D.
Robert Burgess, a professor at The Jackson Laboratory in Bar Harbor, Maine, has been awarded an MDA research grant totaling $300,000 over three years. Burgess and co-investigator Scott Harper, associate professor at Nationwide Children’s Hospital Center for Gene Therapy in Columbus, Ohio, will test an AAV gene therapy approach to specifically block the altered form of the GARS gene in a newly developed mouse model for type 2D Charcot-Marie-Tooth disease (CMT). Successful completion of these studies could lead to a new therapy for type 2D CMT and provide a proof of principle for this approach that may be applicable to other types of CMT.
Funding for this MDA research grant began Aug. 1, 2015.
-
SMA - Arthur Burghes, Ph.D.
Grant Announced: Summer 2015Disease Type: Spinal Muscular Atrophy (SMA)“Arthur Burghes is working to develop a new technique that will allow scientists to accurately predict the severity of a ...”
SMA - Arthur Burghes, Ph.D.
Arthur Burghes, professor of biological chemistry and pharmacology, molecular genetics, and neurology at The Ohio State University Wexner Medical Center in Columbus, was awarded an MDA research grant totaling $188,613 over two years to refine how a genotype can be used to predict the severity of spinal muscular atrophy (SMA). Burghes will develop assays to test whether SMA patients have either intact or defective SMN2 genes, which serve as the backup for the SMN1 gene lost in SMA patients. Since the level of SMN2 determines the disease severity in SMA patients, these assays may be able to better predict how disease may progress in an individual.
Funding for this MDA research grant began Aug. 1, 2015.
-
SMA - Arthur Burghes, PhD
Funded: 08/01/19 through 07/31/21Grant Announced: Summer 2019Disease Type: Spinal Muscular Atrophy (SMA)“In this proposal we aim to identify the genes that modify the severity of SMA and the changes in them that cause modific...”
SMA - Arthur Burghes, PhD
“In this proposal we aim to identify the genes that modify the severity of SMA and the changes in them that cause modification of severity. The identification of these changes will improve the ability to predict severity with a DNA test and initiate treatment at the appropriate time. In addition, these modifier genes can be novel therapeutic targets that can act independently of current therapies to increase SMN.”
Arthur Burghes, PhD, professor of Biological Chemistry and Pharmacology, Molecular Genetics, and Neurology at The Ohio State University Wexner Medical Center in Columbus, was awarded an MDA research grant totaling $200,000 over two years to study genes that might have the potential to modify the severity of spinal muscular atrophy (SMA).
In SMA, mutations in the survival motor neuron 1 gene (SMN1) prevent a person from making enough survival motor neuron protein (SMN). Lack of this protein is what causes the loss of motor neurons, muscle weakness, and paralysis that define SMA. In addition to the SMN1 gene, there is also a related SMN2 gene, which functions as a “backup” to producing some amount of SMN protein.
Dr. Burghes was previously awarded MDA funding to study how SMN2 copy number might predict severity of SMA. Currently SMN2 copy number is the best predictor of SMA severity and in general, having more SMN2 copies results in less severe SMA. However, both SMA type 2 and type 3 patients have three copies of SMN2. In this new work, Dr. Burghes will study the DNA of siblings who have three copies of SMN2, but one has SMA type 2 while the other has SMA type 3. He aims to identify new genes with the potential to modify the severity of the symptoms of SMA, which may potentially become novel therapeutic targets.
-
DMD/BMD — Dean Burkin, Ph.D.
Grant Announced: Summer 2012Disease Type: Becker Muscular Dystrophy (BMD)“Burkin is working to test the hypothesis that laminin-111 protein therapy can prevent muscle damage after disease onset ...”
DMD/BMD — Dean Burkin, Ph.D.
Dean Burkin, associate professor of pharmacology at the University of Nevada School of Medicine in Reno, was awarded an MDA research grant totaling $308,028 over three years to study laminin-111 protein therapy for Duchenne (DMD) and Becker (BMD) muscular dystrophies.
Burkin and colleagues have shown in previous work that systemic delivery of the laminin-111 protein works as a potent therapeutic in a mouse model of DMD. Laminin-111 treatment in these mice results in stabilization of the muscle membrane and a reduction in levels of an enzyme called creatine kinase, or CK, which is a sign of muscle damage.
It remains unclear, however, whether laminin-111 protein therapy in people with DMD may be able to prevent muscle damage, or reverse it after it's already started.
Burkin is working to test the hypothesis that laminin-111 protein therapy can prevent muscle damage after DMD disease onset in mouse models of DMD and in dogs that have the disease.
To do this, Burkin and colleagues first plan to determine if laminin-111 prevents muscle damage after disease onset in the two animal models. Next, the team will test to see whether laminin-111 prevents heart muscle deterioration (cardiomyopathy) in mouse models of DMD, and finally the group will study whether laminin-111 prevents muscle disease in dogs with DMD.
Favorable results could pave the way for development of laminin-111 as a therapeutic for DMD and BMD.
Funding for this MDA grant began Aug. 1, 2012.
-
DMD – Dean Burkin, Ph.D.
Funded: 08/01/17 through 07/31/20Grant Announced: Summer 2017Disease Type: Duchenne Muscular Dystrophy (DMD)“I have received other MDA grants which have been critical to these studies,” Dean Burkin says. “This new grant will ...”
DMD – Dean Burkin, Ph.D.
“I have received other MDA grants which have been critical to these studies,” Dean Burkin says. “This new grant will allow us to move an exciting new integrin-targeting small molecule closer to clinical trials for Duchenne muscular dystrophy patients.”
Dean Burkin, professor of pharmacology and directory of cellular and molecular pharmacology and physiology graduate program at the University of Nevada School of Medicine in Reno, was awarded an MDA research grant totaling $300,000 over three years to test the effects of an existing FDA-approved drug on the function of heart and skeletal muscle in a mouse model of Duchenne muscular dystrophy (DMD).
DMD is caused by mutations in the DMD gene, which encodes the dystrophin protein. Dystrophin forms a molecular “glue” that binds muscle cells together and provides structural integrity, and its loss or deficiency in DMD causes progressive muscle weakness and results in premature death.
A second molecular glue, a protein called alpha7beta1 integrin, is also present in muscle, and studies in transgenic mouse models have shown that increased levels of alpha7beta1 can act as a surrogate for the absence of dystrophin and prevent disease progression.
In collaboration with the National Center for Advancing Translational Sciences (NCATS) at the National Institutes of Health (NIH), Burkin and colleagues conducted a muscle cell-based test and screened more than 400,000 compounds to identify drugs that could enhance the activity of alpha7beta1. They identified a novel compound called SU9516 that increases levels of alpha7beta1 integrin protein and prevents muscle disease in the mdx mouse model of DMD.
SU9516 has an FDA-approved analog called sunitinib, and Burkin’s team will now test the effectiveness of this drug in mouse models of DMD. Since sunitinib already is FDA-approved (to treat some forms of cancer), successful outcomes of this study could lead to future clinical trials and rapid translation into a novel class of integrin-based therapeutics for DMD.
-
CMD - Dean Burkin, PhD
Funded: 08/01/19 through 07/31/22Grant Announced: Summer 2019Disease Type: Congenital Muscular Dystrophy (CMD)“We hope this laminin-enhancing small molecule will slow muscle pathology and improve muscle strength in a mouse model of...”
CMD - Dean Burkin, PhD
“We hope this laminin-enhancing small molecule will slow muscle pathology and improve muscle strength in a mouse model of laminin-alpha2-related congenital muscular dystrophy. Successful preclinical outcomes from this study will form the basis for potential future clinical trials for patients with this disease.”
Dean Burkin, PhD, professor of Pharmacology at the University of Nevada School of Medicine in Reno, was awarded an MDA research grant totaling $300,000 over three years to study a laminin-enhancing small molecule for treating congenital muscular dystrophy (CMD).
Laminin-alpha2-related congenital muscular dystrophy, also known as merosin-deficient congenital muscular dystrophy (MDC1A), is a muscle disease with no cure or treatment. Children with MDC1A typically die at a young age. MDC1A is caused by mutations in the laminin-alpha2 gene, which lead to a lack of merosin or laminin-211/221 in muscle. Laminin-511 (aka, laminin-alpha5) is another form of laminin found in the muscle of MDC1A patients and is similar in structure and function to laminin-211.
Dr. Burkin has recently identified an FDA-approved small molecule, designated laminin-enhancing small molecule 5 (LEM5), that increases laminin-alpha5 in cultured skeletal muscle cells and improves muscle pathology and function in a mouse model of MDC1A. With this new funding, Dr. Burkin will use a LEM5 analog (a modified version of the FDA-approved drug) to determine optimal dose, safety, and efficacy in reducing symptoms in MDC1A mice.
-
Establishing a network of highly trained and reliable CMT clinical evaluators – Joshua Burns, PhD
Funded: 09/01/21 through 08/31/22Grant Announced: 2021Disease Type: Charcot-Marie-Tooth Disease (CMT)“Charcot-Marie-Tooth disease (CMT) is a genetic neuromuscular disease, causing muscle weakness, foot deformities, and fre...”
Establishing a network of highly trained and reliable CMT clinical evaluators – Joshua Burns, PhD
"Charcot-Marie-Tooth disease (CMT) is a genetic neuromuscular disease, causing muscle weakness, foot deformities, and frequent trips and falls or difficulty walking. There is currently no cure for CMT, but promising treatments are on the horizon."
Joshua Burns, Ph.D., is a Professor of Paediatric Neuromuscular Rehabilitation and Associate Dean Research at the University of Sydney in Sydney, NSW, Australia, has been awarded an MDA Idea Grant totaling $25,000 for one year to develop training and resources and accreditation program for clinical evaluators worldwide to help assess patients with CMT.
A robust clinical outcome assessment (COA) is available online, www.clinicaloutcomemeasures.org, for use with individuals with CMT. The online has over 200 clinical evaluators registered worldwide. To avoid evaluator drift and variability in assessment measurements, online training and accreditation systems are significant especially during the COVID-19 era. Dr. Burns's project aims to make eHealth training and resources available and accredit clinicians to use COAs as a reliable measurement for clinical trials and how to use it accurately.
-
Uncovering the Genomic Architecture of Undiagnosed Neuromuscular Disease – Daniel Calame, MD, PhD
Funded: 09/01/21 through 08/31/24Grant Announced: 2021Disease Type: Congenital Muscular Dystrophy (CMD)“Neuromuscular diseases result from dysfunction in the spinal cord, nerves or muscles. This dysfunction is often caused b...”
Uncovering the Genomic Architecture of Undiagnosed Neuromuscular Disease – Daniel Calame, MD, PhD
"Neuromuscular diseases result from dysfunction in the spinal cord, nerves or muscles. This dysfunction is often caused by mutations in our genes. Genetic advances have resulted in the discovery of more than 500 genes which cause neuromuscular diseases."
Daniel Calame, M.D., Ph.D., is a Postdoctoral Fellow at Baylor College of Medicine in Houston, TX, has been awarded an MDA Development Grant totaling $220,000 for three years to improve rates of diagnoses expand the knowledge in neuromuscular genetics using RNA-seq and long-read whole genome sequencing.
Despite these advances, more than half of patients with neuromuscular disease still lack a genetic diagnosis. Several factors contribute to low diagnostic rates. Utilization of RNA-seq and long-read whole genome sequencing in a cohort of well-characterized patients with undiagnosed neuromuscular disease will help discover novel mechanisms of neuromuscular disease and create opportunities for new treatments.
-
DMD/BMD — Michele Calos
Grant Announced: Summer 2010Disease Type: Duchenne Muscular Dystrophy (DMD)“Pictured: cross-section of a normal mouse leg muscle that has been stained for dystrophin, the protein missing in Duchen...”
DMD/BMD — Michele Calos
Michele Calos, professor in the department of genetics at Stanford University School of Medicine in Stanford, Calif., received a $200,000 MDA research grant to develop a new stem cell-based therapy for Duchenne muscular dystrophy (DMD).
"Funding from MDA is critical for us to develop these new ideas that could lead to novel strategies to combat DMD," Calos said. "A prior MDA grant has allowed us to make progress in the studies, and we are now poised to perform the critical experiments."
The new work involves extraction of stem cells from DMD mice. A good copy of the dystrophin gene, necessary but missing in DMD, will be added to the cells, which will then be returned to the mouse where they are expected to home in on damaged muscle and help create new muscle fibers to replace those damaged by the disease. The investigative team will also conduct experiments using a type of adult human stem cell called "adipose-derived mesenchymal stem cells," which are purified from fat tissue and have been shown to develop into muscle.
Successful results in these experiments eventually may lead to clinical trials and may indicate potential for use of a similar approach in other muscle diseases.
"Progress in the development of stem cell therapies offers major hope for regeneration of muscle mass and strength," Calos said, even for those in whom the disease has progressed.
Funding for this MDA grant began August 1, 2010.
-
LGMD – Michele Calos, Ph.D.
Grant Announced: Summer 2016Disease Type: Limb-Girdle Muscular Dystrophy (LGMD)““We are inspired to study LGMD2B and 2D because we are working with families who have patients with these disorders,...”
LGMD – Michele Calos, Ph.D.
Michele Calos, professor of genetics at Stanford University School of Medicine in California, was awarded an MDA research grant totaling $300,000 over a period of three years to test gene therapy approaches in mouse models of limb-girdle muscular dystrophy (LGMD) types 2B and 2D.
With colleagues, Calos will study a gene therapy approach based on delivering the therapeutic gene — either dysferlin or alpha-sarcoglycan — through the bloodstream. The DNA will be injected rapidly into a vein near the ankle, where it will then be carried by the blood to all leg muscles.
The goal of this work is to demonstrate effective DNA delivery that results in improved condition of muscles and muscle function in the mice. A sequence-specific integration system will specifically target the therapeutic gene into a safe and permanent section of the genome. Because the gene will become a permanent part of the chromosomes in muscle cells, the therapy will act long-term.
These studies will pave the way for larger animal studies and human clinical trials. Success in LGMD types 2B and 2D may demonstrate that a similar approach will work for all 10 forms of recessive limb-girdle muscular dystrophy. Furthermore, this type of approach also may be helpful for other neuromuscular diseases.
Funding for this MDA research grant began Aug. 1, 2016.
-
CMD — Kevin Campbell, Ph.D.
Grant Announced: Summer 2012Disease Type: Congenital Muscular Dystrophy (CMD)“Longtime MDA grantee Kevin Campbell says current research aims to identify proteins that contribute to the initiation of...”
CMD — Kevin Campbell, Ph.D.
MDA awarded a research grant totaling $375,000 over three years to Kevin Campbell, professor of neurology and internal medicine at the University of Iowa in Iowa City. The funds will help support Campbell's study of a process called protein O-mannosylation in a mouse model of congenital muscular dystrophy (CMD).
O-mannosylation is an important protein modification that occurs in mammals. Diseases caused by O-mannosylation defects include severe muscular dystrophies characterized by profound muscle weakness as well as central nervous system impairment. Despite extensive efforts, the genetic causes of most occurrences of these diseases remain unknown.
The only known O-mannosylated protein is alpha-dystroglycan, which works in skeletal muscle to link connective tissue (the extracellular matrix) with the cellular framework or "scaffolding" known as the cytoskeleton.
Campbell aims to identify proteins that play a role in initiating O-mannosylation and then investigate their function in cell culture models. He also is working to generate a new O-mannosylation-deficient mouse model in which to study any newly identified proteins.
There is a critical need for better understanding of the mechanism responsible for this modification to develop new treatment options for O-mannosylation deficient disease, Campbell says. "Besides direct patient health benefits, identification of new players involved in protein O-mannosylation will open new avenues to understand O-mannosylation deficient muscular dystrophies."
Funding for this MDA grant began Aug. 1, 2012.
-
Periodic Paralysis — Stephen Cannon, M.D., Ph.D.
Grant Announced: Summer 2011Disease Type: Ion Channel Diseases““With years of support,” Cannon said, “our MDA-sponsored work progressed from establishing the functional abnormal...”
Periodic Paralysis — Stephen Cannon, M.D., Ph.D.
MDA has awarded a research grant totaling $404,274 over three years to Stephen Cannon, professor of neurology and associate dean for undergraduate medical education at the University of Texas Southwestern Medical Center in Dallas. The funds will help support Cannon’s research into attacks of paralysis in people with periodic paralysis (PP).
Periodic paralysis is a muscle disorder in which affected individuals have transient attacks of muscle weakness lasting hours to days.
With prior support from MDA, Cannon and colleagues generated a mouse model for hypokalemic periodic paralysis, in which attacks are triggered by low blood potassium levels. Using the new mouse model, the team intends to establish how shifts in potassium level, exercise and stress trigger attacks of weakness.
Preliminary work already has identified several strategies to reduce the risk of an attack or to reduce the severity of weakness, through the use of available drugs or by modifying the level of muscular activity.
“Support from the MDA has been a vital component of our research program on myotonia and periodic paralysis for two decades,” Cannon said. “MDA’s commitment to funding investigation into the fundamental biology of these diseases has yielded many important advances.
“Through the generosity of donors and the wisdom of the MDA Board of Directors, important work on muscle disease has been able to not only continue, but also to flourish.”
Funding for this MDA grant began August 1, 2011.
-
Periodic Paralysis - Steve Cannon, M.D., Ph.D.
Grant Announced: Winter 2016Disease Type: Endocrine Myopathies“Steve Cannon is investigating the mechanistic basis for exercise-induced weakness in periodic paralysis.”
Periodic Paralysis - Steve Cannon, M.D., Ph.D.
Steve Cannon, professor of physiology at the David Geffen School of Medicine at the University of California, Los Angeles, was awarded an MDA research grant totaling $300,000 over three years to investigate a new hypothesis about the underlying mechanisms involved in periodic paralysis. New preliminary studies in research models have revealed a profound loss of muscle force within minutes of recovery from exposure to high carbon dioxide levels, which Cannon and colleagues suggest may be a surrogate for the exercise-induced attacks of weakness that occurs in patients. Cannon and his team will investigate the mechanistic basis for exercise-induced weakness in periodic paralysis, will strategically select drugs that may block this process, and will test the potential of these drugs to foreshorten or prevent attacks of periodic paralysis.
Funding for this MDA research grant began Feb. 1, 2016.
-
Pompe and LGMD – Dr. Samya Chakravorty, PhD
Funded: 08/01/18 through 07/31/21Grant Announced: Summer 2018Disease Type: Limb-Girdle Muscular Dystrophy (LGMD)“This work will specify what proportion of LGMD multi-genic cases can reach a molecular diagnosis and get enrolled in cli...”
Pompe and LGMD – Dr. Samya Chakravorty, PhD
“This work will specify what proportion of LGMD multi-genic cases can reach a molecular diagnosis and get enrolled in clinical trials. Besides the clinical impact, this project will also have a major impact on our basic scientific understanding of how different LGMD subtypes and related diseases such as Pompe disease are related in the muscle genome.”
Dr. Samya Chakravorty, postdoctoral research fellow at Emory University in Atlanta, Georgia, was awarded an MDA Development Grant totaling $180,000 over 3 years to study the functional resolution of multi-genic Pompe and limb-girdle muscular dystrophy (LGMD).
LGMD subtypes are a group of genetic disorders with overlapping phenotypes among themselves and with Pompe disease, complicating accurate disease diagnosis. Genetic testing can aid in this process, but some estimates indicate that even after extensive genetic testing, 30% to 40% of neuromuscular disease patients never receive a complete molecular diagnosis. This is likely due to a combination of new disease genes not identified, as well as a combination of genes that can contribute to disease manifestation. Combinatorial functional assays will confirm this finding and reveal how gene networks may impact disease manifestations, which will improve molecular diagnosis.
Dr. Chakravorty and colleagues plan to use a discovery-driven approach to correlate gene networks with disease phenotype using patient muscle biopsy tissue to test the hypothesis that complex genetic pathways contribute to the Pompe and LGMD mechanism with high prevalence, potentially by synergistic effects of multiple genes.
-
FSHD — Joel Chamberlain, Ph.D.
Grant Announced: Summer 2012Disease Type: Facioscapulohumeral Muscular Dystrophy (FSH, FSHD)“Many years of research have led recently to an understanding of the unique series of molecular events necessary to cause...”
FSHD — Joel Chamberlain, Ph.D.
Joel Chamberlain, research assistant professor in the department of medicine at the University of Washington in Seattle, was awarded an MDA research grant totaling $330,780 over three years to study a therapeutic approach called RNA interference (RNAi) for the treatment of facioscapulohumeral muscular dystrophy (FSHD).
"Despite the surprising complexity of the FSHD disease pathway, several lines of research converge to identify DUX4 protein production as the final step in the disease pathway," Chamberlain says.
Now, Chamberlain and colleagues are working to develop a therapy that targets production of DUX4 through interaction with DUX4 messenger RNA, or mRNA. (RNA is a chemical cousin to DNA and is involved in a chain of steps between DNA and cellular protein production.)
"To carry out RNAi, we will use an RNA that specifically binds to the DUX4 mRNA and directs the cell to destroy the target DUX4 mRNA," Chamberlain explains. "Once inside the muscle, the RNA made to carry out RNAi is produced continuously to treat disease and is expected to remain active for many years."
The team also is working to create a mouse model of FSHD that mimics the human disease. The investigators will use the model — as well as new cell and mouse models from collaborating laboratories — to test their DUX4 RNAi approach. They plan to make the model available to other researchers for the testing of additional potential therapies.
"We have developed this targeted approach, referred to as RNA interferenceor RNAi, to reverse disease," Chamberlain says. "It works well in mouse models of muscular dystrophies, and we are the only laboratory to have succeeded in changing the course of the disease by delivering this therapy to all the muscles of the mouse with a single intravascular (into a blood vessel) injection."
Funding for this MDA grant began Aug. 1, 2012.
-
Generation of large dystrophins in muscle via AAV vector delivery - Jeffrey Chamberlain, PhD
Funded: 05/01/20 through 04/30/22Grant Announced: Summer 2020Disease Type: Duchenne Muscular Dystrophy (DMD)“Our new approach will continue development of a novel approach to delivering larger, possibly up to full-length, dystrop...”
Generation of large dystrophins in muscle via AAV vector delivery - Jeffrey Chamberlain, PhD
"Our new approach will continue development of a novel approach to delivering larger, possibly up to full-length, dystrophins."
Jeffrey Chamberlain, Ph.D., Professor and McCaw Endowed Chair of Muscular Dystrophy at the University of Washington School of Medicine, was awarded an MDA restricted research grant totaling $471,693 over two years to develop improved dystrophin vectors that can deliver larger and more functional dystrophins to muscles throughout the body in patients with Duchenne muscular dystrophy (DMD).
Current approaches for DMD gene therapy rely on AAV delivery of micro-dystrophin or gene editing tools, which each produce smaller than normal dystrophins. Dr. Chamberlain plans to develop improved dystrophin vectors than can produce larger and more functional dystrophin to muscles of DMD patients. If successful, this technology would be applicable to any DMD or Becker’s muscular dystrophy (BMD) patient.
-
FSHD – Joel Chamberlain, Ph.D.
Grant Announced: Summer 2016Disease Type: Facioscapulohumeral Muscular Dystrophy (FSH, FSHD)““Our area of focus is important in achieving a greater understanding of the early changes in muscle resulting from DUX...”
FSHD – Joel Chamberlain, Ph.D.
Joel Chamberlain, research associate professor at the University of Washington School of Medicine in Seattle, was awarded an MDA research grant totaling $300,000 over a period of three years to increase understanding of the role of DUX4 protein in facioscapulohumeral muscular dystrophy (FSHD).
In the muscle cells of people with FSHD, too much of a protein called DUX4 is made, leading to cell death. To determine how excess DUX4 results in muscle damage in FSHD, Chamberlain and colleagues will examine DUX4 protein production in a range of human biopsy samples in an effort to determine the cellular location of DUX4 and the protein’s relationship to disease-related changes in the muscle.
In parallel, in studies conducted in a unique mouse model of FSHD developed in Chamberlain’s lab, the team also will examine the toxic effects of expressing very low levels of the DUX4 protein. These studies will help assess how DUX4 initiates a chain of events that cause the slow, progressive muscle damage seen in FSHD.
Chamberlain’s work may reveal drug targets and inform the development and testing of therapies for FSHD.
Funding for this MDA research grant began Aug. 1, 2016.
-
DMD/BMD — Jeffrey Chamberlain, Ph.D.
Grant Announced: Additional Grants 2013Disease Type: Duchenne Muscular Dystrophy (DMD)
DMD/BMD — Jeffrey Chamberlain, Ph.D.
Jeffrey Chamberlain, a molecular biologist at the University of Washington, Seattle, was awarded an MDA grant of $328,628 over one year (through Jan. 31, 2014) to develop gene therapy delivery vehicles ("vectors") for gene transfer in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). Chamberlain will develop gene transfer vectors derived from adeno-associated viruses (AAVs). They'll be designed to carry miniaturized versions of the gene for the dystrophin protein, which is missing in DMD-affected muscles and deficient in muscles affected by BMD. The studies are directly related to moving gene therapy for DMD/BMD into clinical trials.
-
MM - David Chan, M.D., Ph.D.
Grant Announced: Spring 2014Disease Type: Mitochondrial Myopathies (MM)“David Chan will study defects in the behavior of mitochondria, which produce energy in cells, in mice with mitochondrial...”
MM - David Chan, M.D., Ph.D.
David Chan, a professor of biology at the California Institute of Technology in Pasadena, has been awarded an MDA research grant totaling $253,800 over three years, to study mitochondria in mice. Mitochondria are the energy-producing parts of cells, and an aspect of their normal functioning involves fusing together and dividing. Chan and colleagues will study mice with defects in the fusion or division of mitochondria to improve the understanding of mitochondrial myopathies.
Funding for this MDA research grant began May 1, 2014.
-
Vijayendran Chandran, Ph.D.
Grant Announced: Winter 2017Disease Type: Friedreich's Ataxia (FA)““Molecular biomarkers are important tools in assessing clinical trial outcomes, measuring disease progression, and eva...”
Vijayendran Chandran, Ph.D.
Vijayendran Chandran, assistant professor in the department of pediatrics, University of Florida School of Medicine in Gainesville, was awarded an MDA research grant totaling $300,000 over three years to identify biomarkers in Friedreich’s ataxia (FA).
FA is caused by deficiency of the frataxin protein. With colleagues, Chandran has developed a mouse model for FA, that exhibits symptoms similar to those seen in people with the disease and in which he can control the onset and progression of disease.
Now the team will use the new model to identify biomarkers at different stages of disease that could predict disease onset and progression in humans.
If successful, Chandran’s work could help clinicians and researchers measure and predict disease progression, and aid in preclinical development and testing of potential FA treatments.
Funding for this MDA research grant began Feb. 1, 2017.
- ‹ previous
- 2 of 13
- next ›