The Pitt Hopkins Research Foundation is thrilled to announce our 2016 Grant Awardees.  Three grants were given at 75K each by the PHRF, and two grants were given through the University of Pennsylvania, Million Dollar Bike Ride, at 50K each.  A total of $325,000 was awarded for Pitt Hopkins research this year:  

Tilly Hadlow Young Investigator Award:

Joseph Alaimo, Ph.D., Principal Investigator; Sarah Elsea, Ph.D., Co-Investigator/Mentor, Baylor College of Medicine, Awarded $75,000, PHRF

Delineating Therapeutic Targets using Global Metabolic Profiling in Pitt Hopkins syndrome:  Defining the cellular defects due to alterations in TCF4 function is paramount in order to determine the proper molecular and biochemical targets for therapeutic intervention in Pitt-Hopkins syndrome. To identify and characterize the biochemical and molecular dysfunction due to altered TCF4 function, we plan to take a clinical and translational approach by recruiting a cohort of individuals with PTHS in collaboration with the Pitt-Hopkins Research Foundation and current PTHS clinics, phenotypically and molecularly characterizing the cohort, and employing state-of-the-art metabolomics screening to identify pharmacologically targetable molecular and biochemical pathways. Our unique approach will utilize a special type of biochemical genetic test called global metabolomics assisted pathway screening (Global MAPS). Global MAPS is currently the most comprehensive small molecule screen available in the clinical setting and is only available through Baylor College of Medicine’s Biochemical Genetics Diagnostic Laboratory. Global MAPS surveys greater than 1000 small molecules in human plasma, pinpointing defects in pathways unmeasurable by standard clinical testing methods, offering a comprehensive and in-depth analysis of patient samples and metabolic status. Our overall goal is to employ Global MAPS analysis in patients with PTHS to identify novel pathway alterations and to understand the basis of TCF4 function in cells. In addition, our proposal will serve as a functional confirmation of current molecular findings in PTHS research, including RNA-sequencing and gene expression profiling, thereby refining the molecular and biochemical targets that would benefit most from therapeutic intervention. Our novel but complementary approach will promote additional analysis to identify points of data convergence among other research groups thereby expediting the process toward targeted therapeutic intervention and clinical trials. Any Pitt-Hopkins families interested in participating in metabolomics profiling are highly encouraged to contact either Drs. Sarah Elsea or Joseph Alaimo at Baylor College of Medicine using the following information: email: Alaimo@bcm.edu   Phone: 832-824-8936

Benjamin D. Philpot, Ph.D., Principal Investigator; Alexander D. Kloth, Ph.D., Co-Principal Investigator; Courtney L. Thaxton, Ph.D., Co-Principal Investigator, The University of North Carolina at Chapel Hill, Awarded $75,000, PHRF

Characterization and Generation of PTHS Model Mice for Rational Therapeutic Discovery:  Pitt-Hopkins syndrome (PTHS) is a rare neurodevelopmental disorder characterized by intellectual disability, absent speech, seizures, ataxia, and breathing anomalies. In support for future therapeutic development for PTHS, we will pursue two independent aims: (1) to uncover the neural impairments that are common across multiple PTHS mouse models, and (2) to develop new tools to analyze TCF4 expression in neuronal subtypes throughout development and adulthood. In the first aim, we will follow up on our finding that long-term changes in synaptic function related to experience are enhanced in multiple PTHS-related mouse models. We hypothesize that this deficit is related to altered function of a glutamate receptor, the NMDA receptor, and we will rigorously test this hypothesis using electrophysiology, biochemistry and pharmacological methods in multiple PTHS-related mouse models. In the second aim, we will develop a unique mouse model toward determining effective drug targets that affect TCF4 expression levels, as well as be able to alter TCF4 activity in a spatiotemporal manner. This novel binary “reporter-reinstatement” mouse will not only allow for a stream-lined and genetically precise approach to drug discovery for PTHS, but also will allow us to determine the most efficacious time in which to reinstate TCF4 function to alleviate the pathophysiologies associated with PTHS. In all, the proposed project pursues incisive approaches that will provide guidance to the development of PTHS therapeutics.

Kindal Kivisto Award for Promising Young Researchers:

Andrew John Kennedy, Ph.D., Principal Investigator; J. David Sweatt, Ph.D., Co-Principal Investigator, Evelyn F. McKnight Brain Institute, The University of Alabama at Birmingham, Awarded $75,000, PHRF

Investigating Therapies for Pitt-Hopkins Syndrome:  The central strategy of our research program consists of two goals: the near-term goal to identify FDA approved drugs as potential translatable therapies for Pitt-Hopkins Syndrome (PTHS) and the long-term goal to develop novel neuroepigenetic therapies that fundamentally reverse the effects of PTHS.  Over the past three years, we have characterized a genetically engineered heterozygous deletion mouse model of PTHS (Tcf4 +/-), validated the histone deacetylase enzyme Hdac2 as a target to treat the cognitive deficits associated with PTHS, and undertaken a drug screening program.  This grant will investigate the efficacy of Fingolimod (trade name Gilenya), as well as other FDA approved therapeutics that target Hdac2, to improve learning, problem solving, and associative memory in PTHS mice.  These experiments will focus on identifying a plausible drug candidate that can be translated to a clinical setting and effectively improve cognition in PTHS patients. Additionally, more advanced epigenetic therapies will be developed to address the genetic cause of PTHS.  Every person has two functioning copies of Tcf4 with the exception of individuals with PTHS, who have a mutation or deletion that yields only one functioning copy.  Epigenetic therapies, which alter the epigenetic states at specific genes within the genome, are being designed to allow PTHS models to use their one functioning copy of Tcf4 twice as much, hopefully restoring full Tcf4 function and reversing the cognitive deficits associated with Pitt-Hopkins.  Taken together, these approaches investigate already-available FDA approved drugs and cutting edge genetic techniques to identify potential therapies that improve cognition in the near-term and attempt to address and compensate for the underlying cause of Pitt-Hopkins Syndrome.

Brady Maher, Ph.D., Principal Investigator; Huei-Ying Chen Ph.D.; Stephanie Cerceo-Page, Ph.D.; Lieber Institute for Brain Development, Johns Hopkins School of Medicine, Awarded $50,000, UPenn, MDBR

Exploring the impact of a TCF4 mutation on the physiology of inhibitory neurons of the prefrontal cortex:  PTHS is a neurodevelopmental disorder due to mutation or deletion of one copy of the TCF4 gene. TCF4 is a transcription factor that can regulate the expression of many downstream genes and therefore regulates the genetic programs necessary for normal brain development. We measured the expression of TCF4 mRNA across the lifespan in humans and rodents and observed a peak in TCF4 expression occurs during the formation of the cerebral cortex, a region of the brain important to higher cognitive functions including learning and memory. Using a mouse model of PTHS that has a mutation in one copy of the TCF4 gene, we observed that TCF expression is blunted during the developmental peak in expression compared to control animals, and we believe this indicates a causal time period for the development of PTHS. Unfortunately, this critical period occurs in utero and prior to when diagnosis is currently made, thus complicating our ability design treatment strategies during this causal phase of the disorder. Therefore, our research group is focused on understanding the underlying pathophysiology that produces symptomatology in PTHS so that we can normalize this pathophysiology in children and adults. Using our animal models of PTHS, we have identified a sodium channel that is normally expressed in the peripheral nervous system, but is ectopically expressed in the central nervous system when TCF4 is mutated. Experiments are currently underway to determine if blocking this Na channel with drugs can lead to improvement on behavioral tests in our PTHS mouse model. In our current proposal, we would like to follow up a preliminary result that suggests inhibitory transmission onto excitatory neurons in the cortex is decreased in the PTHS mouse compared to control littermates. In addition, using RNA sequencing of the PTHS mouse model we observed that many genes that are specific to inhibitory neurons show decreased expression compared to control animals, and we identified a specific population of inhibitory neurons (cortistatin positive) that normally show high levels of TCF4 expression. These cortistatin positive interneurons are known to release a neuropeptide called cortistatin that has been shown to inhibit the generation of seizures and regulate sleep states. Given the prevalence of seizures and sleep disturbances in PTHS, we believe this population of inhibitory neurons may underlie clinical aspects of the disorder. Therefore, we propose to breed the PTHS mouse with another mouse that allows us to visualize cortistatin positive interneurons and we will use electrophysiology and microscopic imaging to determine if these cells are disrupted in the PTHS mouse model compared to control littermates. If deficits are observed in this population we will determine the cellular and molecular mechanism using pharmacological rescue and/or molecular phenocopy. Identified molecular mechanisms will then be deemed potential therapeutic targets and these targets will be tested for their ability to normalization of behavioral deficits in the PTHS mouse.

Tõnis Timmusk, Ph.D., Principal Investigator; Mari Sepp, Ph.D., Co-Investigator, Tallinn University of Technology, Estonia, Awarded $50,000, UPenn, MDBR

Regulation of TCF4 transcriptional activity in neurons:  Transcription factor TCF4 (alias ITF2, SEF2 or E2-2) is a broadly expressed protein involved in the development and functioning of many different cell types. Recent studies point to important roles for TCF4 in the nervous system. Specifically, human TCF4 gene is implicated in susceptibility to schizophrenia and mutations in TCF4 cause Pitt-Hopkins syndrome (PTHS), a rare developmental disorder characterized by severe motor and mental retardation, typical facial features and breathing anomalies. The mutation may be in different parts of the gene, but it appears in only one allele. Whereas in many other genes the other, unaffected allele would be able to compensate for the defect, this is not the case in TCF4. This indicates that the protein encoded by the TCF4 gene is essential for the development of the nervous system, and that human development depends significantly on the amount of this protein in the brain and body. Our previous data have suggested that synaptic activation of nerve cells, that is the basis of brain function, leads to activation and phosphorylation of TCF4 protein. Phosphorylation is the addition of a phosphate group to a protein or other organic molecule. Phosphorylation turns many proteins on and off, thereby altering their function and activity. The current project is aimed to find out how the activity and phosphorylation of TCF4 protein is regulated inside nerve cells of the brain, and to characterize the phosphorylation pattern of activated TCF4. Additionally, we want to determine which genes are targeted by TCF4 in nerve cells after synaptic activation. Since Pitt-Hopkins syndrome manifests itself at an early stage, there are better chances for its treatment due to the greater plasticity of children’s brains. Increasing the amount and/or activity of the functional TCF4 protein produced from the healthy allele is among possible approaches to develop drugs for Pitt-Hopkins syndrome treatment. We believe that our project could lead to the discovery of novel possibilities for increasing the activity of TCF4 in nerve cells that could be useful to develop treatments for therapeutic intervention of Pitt-Hopkins syndrome.

 

This year, The PHRF is deeply moved to award the “Kindal Kivisto Award for Promising Young Researchers,” in honor of Kindal Kivisto a young woman who died far too soon, at the age of 29 this past October. Kindal did not have Pitt Hopkins, but was close friends with the Sandiford family whose daughter, Finley was diagnosed in 2014. In light of this, her family asked that all donations at the funeral be made to the Pitt Hopkins Research Foundation. The outpouring of love and support for this special young woman was overwhelming.  A total of $25,000 was donated to the Pitt Hopkins Research Foundation in memory of Kindal.  Her family wrote to us during this time, and we will never forget them saying that hoped some good could come of this tragedy.

So this year, in honor of Kindal and her family, we are awarding a grant to Dr. Andrew Kennedy of the The Sweatt Lab at The University of Alabama, Birmingham in her name, with the knowledge that the promise taken from her may in some small way be countered by the incredible hope and promise this young researcher creates for our families every day.

Kindal and Family… Thank you for making this possible for our children. You will not be forgotten.

2015: A YEAR OF INCREDIBLE GROWTH

FOR THE PITT HOPKINS RESEARCH FOUNDATION!

Thanks to everyone who helped make 2015 our best year yet. Let’s make 2016 even better!

2015 Fundraising totals are in, and they are Bigger Than EVER:  $460,000

 

That’s $60 K more than last year and, this year we didn’t even have those amazing EASTSIDE high school kids (who brought in $130K) helping! So that means, as a group we brought in nearly $200K more than last year. More and more new families joined the effort and with a 75 percent increase in family involvement over last year!

2015 Accomplishments

There is a tremendous amount going on at PHRF; we are now supporting 7 research projects. When we started, just 3 years ago, there was virtually no laboratory research being done on this exceedingly rare disorder. This growth means more scientists are focusing on Pitt Hopkins; more labs are working together and sharing knowledge, ideas, and resources; and we are spurring a range of approaches to developing treatments and a cure for Pitt Hopkins. In short, it means we have more scientific momentum than ever towards what we all want—to change the lives of those who live every moment of every day with the debilitating symptoms of Pitt Hopkins.

Here are a few specifics on progress we have made this year in several key areas:

1. Reversal: Reversal projects target the underlying cause of Pitt Hopkins and have the greatest likelihood of having a profound impact on symptoms. Our lab at the University of Alabama Birmingham (UAB) has found a drug therapy that reverses the symptoms of Pitt Hopkins in mice! There is a similar, less toxic drug, currently being tested in the mice that may be contender for trials in the near future.

2. Treatment: Treatment projects seek to improve the quality of life for those with Pitt Hopkins by addressing one or more symptoms. This year the PHRF supported two studies at top labs (the Powell Lab at University of Texas Southwestern and the Katz Lab at Case Western University) looking at epilepsy and breathing issues. Currently our mouse models are being tested for seizure activity and hyperventilation. Once these studies are done we will know whether or not we have a legitimate target for testing treatment-centered therapeutics in the mice.

The Katz Lab at Case Western also studies breathing issues in Rett Syndrome mice, and they have brought a drug called Ketamine to trial this year in Rett Syndrome. This may be a possible target drug for our children as well. It’s too early for results on these trials, but we will share news as soon as we get them.

3. Clinics: The PHRF launched two dedicated Pitt Hopkins Clinics, one at UCSF in San Francisco and one at UT Southwestern in Dallas. We are also working with the National Institute of Mental Health to launch a Pitt Hopkins study site (Summer, 2016) where children from all over the world will be able to be seen at no cost. We hope to continue building this network of clinics around the world where doctors will share and update each other regularly on the latest and most effective medical practices for our children. These clinics and study sites will also be essential in helping develop a comprehensive natural history of Pitt Hopkins, a must for moving forward with clinical trials.

4. Basic science: To inform all our approaches for treating and hopefully curing Pitt Hopkins Syndrome, we must continue expanding our understanding of the neurobiology behind PTHS and the mutated Tcf4 gene that causes it. Our funded labs are doing exactly this and have made several discoveries, including the finding of dysregulated glutamate receptors. This led us to convince our Dallas clinic to try Amantadine, an already FDA approved NMDA receptor antagonist on some patients, and we are seeing some positive results, including increased focus and reduced hyperactivity.

5. Family Support: We held the first ever Science and Family Conference this September in Washington D.C. Families from all over the world joined together for the first time to hear from our scientists and clinics and to meet other members of our global Pitt Hopkins Family. We had no idea the impact this one weekend would make. We laughed, we cried, we cried some more, and we knew at the end of it all we could not wait TWO more years to see our Pitt Family again. We had originally thought the conference would be biannual. But it was clear, that as a fledgling and growing organization, we needed to do it again, as soon as possible.

MARK YOUR CALENDARS FOR
Nov 5-7, 2016 Dallas, Texas!

6. AAC: We often hear from parents that one of the hardest parts of Pitt Hopkins is the lack of communication, particularly when our kids are suffering. This year we dove into learning more about Augmentative Alternative Communication and held our first AAC conference at the Science and Family Conference. Stay tuned for more of these in 2016.

7. Give Rare champions: We were crowned THE 2015 GIVE RARE CHAMPIONS. We raised $23,000 in ONE DAY and won 15K more in matching grants, beating out organizations far bigger than us!

WHO’S IN FOR GIVE RARE 2016? Email us at:
phrf-fundraising@pitthopkins.org

8. Pinterest: We launched a Pinterest page to share ideas on everything from therapy to gluten free and dairy free desserts to what to buy for Pitt Christmas presents. Join us here.

9. Coriell Cell Bank: We launched the Coriell Cell Bank for Pitt Hopkins skin fibroblasts and plasma so that reseachers all over the world will have easier access to skin and blood cells for research. Email Nahid Turan nturan@coriell.org to start the donating process.

10. Registry: We created the global Pitt Hopkins Registry to gather information and help advance research more efficiently and faster. We have over 200 people registered so far! If you haven’t registered yet, you can do so here.

More families than ever were involved in making this the best year yet at the PHRF! If you are interested in getting involved, contact us at phrf-fundraising@pitthopkins.org.