Grantees

Circuitry and Brain-Body Interactions

The circuitry and brain-body interactions theme focuses on basic research aimed at understanding how the circuits that underlie key brain regions are affected in Parkinson’s disease and how they may contribute to disease initiation and progression. These teams will also investigate how communication between the brain and areas outside the brain are affected over the disease course.

Be a Part of Our Mission

We’re enabling science to go further, faster, and at a greater scale. Follow @ASAP_Research across our social media channels and join our mailing list for exciting updates.

Sign up for updates here.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Redefining Parkinson’s Disease Pathophysiology Mechanisms in the Context of Heterogeneous Substantia Nigra Neuron Subtypes

Study Rationale: The motor symptoms of Parkinson’s disease (PD) result from the degeneration of the dopamine-producing neurons in a brain area called the substantia nigra pars compacta (SNc). Recent findings suggest that the SNc is diverse and is comprised of dopamine neurons with distinct properties. How these dopamine neuron “subtypes” contribute to movement, how they are affected in PD, and how they are modified by deep brain stimulation (DBS) remains unknown.

Hypothesis: Team Awatramani will determine whether (a) the SNc is comprised of pro-motor and anti-motor dopamine neuron subtypes and (b) selective loss of pro-motor neurons in PD causes an imbalance in dopamine neuron subtypes that underlies the motor symptoms of PD.

Study Design: Team Awatramani will separate these neurons into their distinct genetic subtypes, which will allow the team to study their specific physiological, anatomical, and functional properties. The team will also determine the molecular and circuit mechanisms underlying the dysfunction of dopamine neurons in a mouse model of PD (LRRK2 model). Additionally, they will explore whether deep brain stimulation of dopamine neuron inputs contributes to the therapeutic efficacy of this treatment.

Impact on Diagnosis: First, Team Awatramani work will identify which dopamine neuron subtypes degenerate and which circuits are dysregulated in PD. This knowledge will be important for understanding the pattern of SNc neuron loss in PD and efficacy of DBS in patients. By using a LRRK2 model, Team Awatramani’s studies also will identify the molecular targets of the hyperactive LRRK2 enzyme, which will be critical for the optimization of LRRK2 inhibitor drugs and their application in patients.

Leadership
Rajeshwar Awatramani, PhD
Coordinating Lead PI

Rajeshwar Awatramani, PhD

Northwestern University
Mark Bevan, PhD
Co-Investigator

Mark Bevan, PhD

Northwestern University
Daniel Dombeck, PhD
Co-Investigator

Daniel Dombeck, PhD

Northwestern University
Thomas Hnasko, PhD
Co-Investigator

Thomas Hnasko, PhD

University of California at San Diego
Loukia Parisiadou, PhD
Co-Investigator

Loukia Parisiadou, PhD

Northwestern University
Amanda Schneeweis
Project Manager

Amanda Schneeweis

Northwestern University

Project Outcomes

Team Awatramani aims to provide a detailed understanding of the diverse dopamine neurons that exist in the substantia nigra in terms of their molecular signatures, unique anatomical circuits, functional contributions to motor behavior, and how this circuitry is disrupted in Parkinson's disease models. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Understanding and Manipulating Cellular and Circuit-Level Vulnerability to Neurodegeneration in Parkinson’s Disease

Study Rationale: Many people with Parkinson’s disease (PD) develop untreatable cognitive symptoms, including problems with attention, decision-making, and dementia at late stages of the disease, due to changes in a crucial part of the brain, the cerebral cortex. Evidence suggests that aggregation of the protein alpha-synuclein in vulnerable nerve cells interferes with their health and damages the cellular networks required for normal brain function. However, the relationships between alpha-synuclein, vulnerable cells, and network activity are not understood. By identifying and understanding the causes and effects of this damage in brain networks, and affected nerve cells and their connections, Team Biederer aims to enable therapies that directly target this disorder.

Hypothesis: Team Biederer hypothesizes that networks of nerve cells in the cortex of the brain become dysfunctional because of damage caused by pathological deposits of the protein alpha-synuclein in vulnerable cells.

Study Design: Team Biederer will assess how alpha-synuclein pathology progressively impairs network function in the brain cortex and identify the features distinguishing vulnerable from resilient cells using innovative technologies, including imaging of activity in the live brain, measurements of attention, profiling of different cell types and their contents, and high-resolution microscopy of neuronal connections. The team will integrate these data using advanced computational methods to design and test cell-specific interventions to restore the function of the disrupted networks. This study will reveal mechanisms of cortical network damage in Parkinson’s disease and will identify the types of nerve cells suited for therapeutic intervention.

Impact on Diagnosis: Diagnostic biomarkers for PD can leverage Team Biederer’s findings of which types of neurons, their connections, and molecular markers are most affected. For treatment, Team Biederer’s work will define novel mechanisms that can directly restore network function by targeting specific types of cortical nerve cells and their connections.

Leadership
Thomas Biederer, PhD
Coordinating Lead PI

Thomas Biederer, PhD

Yale University
Dani Bassett, PhD
Co-Investigator

Dani Bassett, PhD

University of Pennsylvania
Elena Gracheva, PhD
Co-Investigator

Elena Gracheva, PhD

Yale University
Michael Henderson, PhD
Co-Investigator

Michael Henderson, PhD

Van Andel Institute
Michael Higley, MD, PhD
Co-Investigator

Michael Higley, MD, PhD

Yale University
Tina Matos, PhD
Project Manager

Tina Matos, PhD

Yale University

Project Outcomes

Team Biederer's project will gain mechanistic insight into PD pathology linked to progression to dementia. The team will integrate information across molecular, anatomical, and circuit domains, using mathematical modeling, to reveal and manipulate underlying cellular and network vulnerabilities in the cortex. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Circuit Mechanisms for Dopamine Neuron Vulnerability and Resilience in Parkinson’s Disease

Study Rationale: While neurodegeneration in Parkinson’s disease (PD) affects many cells, dopamine neurons are particularly vulnerable, and their loss drives many of the major motor difficulties in PD. To date, the inner workings of the dopamine neurons themselves have been extensively studied to identify sources for this selective vulnerability. However, neurons in the brain are heavily interconnected and interdependent with their surrounding cells and circuitry. Understanding how the “neighborhood” in which dopamine neurons live and function influences their well-being is a critical missing piece in the puzzle of PD.

Hypothesis: Team Calakos hypothesizes that circuit properties of incoming neuronal connections (synapses), surrounding non-neuronal cells (glial cells), and key modulatory cells (those that produce the chemical signal, acetylcholine) contribute to dopamine neuron loss in PD.

Study Design: Team Calakos will evaluate circuit contributions to dopamine neuron dysfunction in PD using state-of-the-art mouse genetic models and patient-derived stem cell models (organoids). Team Calakos members bring unique, specialized expertise that allows the team to isolate and manipulate each of these three components (synapses, glia, and neuromodulators) individually to test its role in dopamine neuron degeneration. In addition to functional manipulations, the team will capture the molecular signatures of the connections dopamine neurons make with each of its “neighbors” to determine which are most disrupted in PD.

Impact on Diagnosis: Recognizing new processes that cause dopamine neuron demise in PD creates new opportunities for intervention. Team Calakos’ bidirectional tests of function may identify not only circuit properties that accelerate disease, but also identify factors that promote resistance to cell death.

Leadership
Nicole Calakos, MD, PhD
Coordinating Lead PI

Nicole Calakos, MD, PhD

Duke University
Cagla Eroglu, PhD
Co-Investigator

Cagla Eroglu, PhD

Duke University
Sergiu Pasca, MD
Co-Investigator

Sergiu Pasca, MD

Stanford University
Scott Soderling, PhD
Co-Investigator

Scott Soderling, PhD

Duke University
Michael Tadross, MD, PhD
Co-Investigator

Michael Tadross, MD, PhD

Duke University
Oula Khoury, PhD
Project Manager

Oula Khoury, PhD

Duke University

Project Outcomes

Team Calakos' collective team efforts will reveal the extent to which circuit components outside of the dopamine neurons themselves can serve as new targets to slow the progression of dopamine neuron death in Parkinson’s disease. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Mapping the Modulatory Landscape Governing Striatal Dopamine Signaling and Its Dysregulation in Parkinson’s Disease

Study Rationale: Nerve cells that produce the brain chemical dopamine die in people with Parkinson’s disease (PD). These nerve cells extend long and thin fibers called axons that release dopamine from thousands of different points, sending signals to other nerve cells in a brain area called the striatum. Many different types of cells and molecules in the striatum can directly control how dopamine is released, but researchers have not yet discovered which ones are the most important and how they are affected in Parkinson’s. By better understanding this cooperation between striatum and the release of dopamine from axons, the team could provide new knowledge toward ways to restore normal function.

Hypothesis: Team Cragg thinks that other molecules in the striatum play a very important role in controlling the release of dopamine, particularly for the types of dopamine axons that are most vulnerable in PD. The team believes that this role is disrupted in the disease and could be targeted to rescue symptoms.

Study Design: Team Cragg’s international team will combine cutting-edge research methods in mice and human cells that allow the team to study the biology behind Parkinson’s. The team will measure dopamine and other signaling molecules in different areas of the striatum and work out what they do. This work will reveal the biological differences between vulnerable and resistant areas. Team Cragg will use this knowledge to study the most promising molecules in mice that develop PD and in cells from people with PD, to then suggest new ways that might fix the problems with dopamine in the disease.

Impact on Diagnosis: Team Cragg’s discoveries will provide knowledge that may help to find new ways of treating Parkinson’s using medicines that target the key signaling molecules in striatum that control dopamine release.

Leadership
Stephanie Cragg, MA, DPhil
Coordinating Lead PI

Stephanie Cragg, MA, DPhil

University of Oxford
Mark Howe, PhD
Co-Investigator

Mark Howe, PhD

Boston University
Peter Magill, DPhil
Co-Investigator

Peter Magill, DPhil

University of Oxford
Konstantinos Meletis, PhD
Co-Investigator

Konstantinos Meletis, PhD

Karolinska Institute
Richard Wade-Martins, MA, DPhil
Co-Investigator

Richard Wade-Martins, MA, DPhil

University of Oxford
Cláudia Mendes, PhD
Project Manager

Cláudia Mendes, PhD

University of Oxford

Project Outcomes

Team Cragg expects their collaboration to unravel the modulators and circuits within striatum that govern dopamine function for vulnerable versus resistant neurons, and the related dysfunction in these key circuits during disease progression, to provide fresh mechanistic rationale for new therapies for Parkinson’s disease. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Dual Role of Neural Activity in Parkinson’s Disease

Study Rationale: Previous work has shown how the loss of dopamine neurons affects brain activity. In this program, Team Edwards will determine how brain activity influences the neurodegeneration that causes Parkinson’s disease (PD). To understand the onset of disease, the team will identify the earliest changes in brain activity and use them to infer the mechanisms involved. Team Edwards will also manipulate activity directly and determine how it interacts with known genes to produce degeneration.

Hypothesis: Team Edwards hypothesizes that abnormalities in neural activity do not simply reflect PD but actually cause the disease. Researchers’ lack of knowledge about the role of neural activity in Parkinson’s makes it difficult to understand how other, identified factors contribute to disease.

Study Design: Team Edwards will use two models of PD, one based on over-expression of the pathogenic protein alpha-synuclein and the other based on a direct increase in activity. The strategy is to (a) identify the earliest events along the pathway to degeneration and to (b) correlate these with the selective vulnerability of particular neurons to PD. These approaches will reveal the processes specifically affected by PD. Team Edwards will also determine how neural activity intersects with factors previously implicated in PD, providing a foundation to understand how they cause degeneration.

Impact on Diagnosis: With greater understanding around the onset of disease, Team Edwards can further investigate how genetic and environmental factors conspire to produce PD. This will open entirely new areas to arrest and prevent the underlying degeneration.

Leadership
Robert Edwards, MD
Coordinating Lead PI

Robert Edwards, MD

University of California at San Francisco
Zayd Khaliq, PhD
Co-Investigator

Zayd Khaliq, PhD

National Institutes of Health
Ken Nakamura, MD, PhD
Co-Investigator

Ken Nakamura, MD, PhD

Gladstone Institutes
Alexandra Nelson, MD, PhD
Co-Investigator

Alexandra Nelson, MD, PhD

University of California at San Francisco
Talia Lerner, PhD
Co-Investigator

Talia Lerner, PhD

Feinberg School of Medicine at Northwestern University
Haru Yamamoto, BA
Project Manager

Haru Yamamoto, BA

University of California at San Francisco

Project Outcomes

By identifying the earliest changes leading to degeneration, the physiology will indicate mechanisms involved in disease onset. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Reconstituting the Lost Nigrostriatal Circuitry in Parkinson’s Disease

Study Rationale: Team Mobley recently discovered that it is possible to generate new neurons to rebuild the damaged neural circuitries in a Parkinson’s disease model. This establishes a new foundation for developing strategies to reverse the disease. Team Mobley’s proposed project will investigate how to change a cell’s identity to encourage it to become a neuron, how to make the right new type of neurons in the brain to rebuild different circuitries, and how to make new neurons that will not become sick again.

Hypothesis: Team Mobley hypothesizes that astrocytes, an abundant population of non-neuronal cells in the brain, store a latent program that allows them to become neurons if the right types of inducing signals are provided.

Study Design: Team Mobley has designed five sets of experiments to address a series of fundamental questions on cell fate determination and reprogramming. The team will analyze individual cells to elucidate key regulatory events responsible for those cells to become neurons. Team Mobley will search for critical genes that make cell fate change less efficient so that the team can improve the reprogramming efficiency by inhibiting the function of those genes. The team will examine reprogramming in different brain regions to test their benefits to both motor and non-motor symptoms and develop strategies to make new and disease-resistant neurons.

Impact on Diagnosis: More information on cellular reprogramming will pave the way to rebuilding the neural circuitries lost to degeneration. If successful, this will lead to the development of a completely new cell replacement therapy against Parkinson’s disease.

Leadership
William Mobley, MD, PhD
Coordinating Lead PI

William Mobley, MD, PhD

University of California at San Diego
Xiang-Dong Fu, PhD
Co-Investigator

Xiang-Dong Fu, PhD

Westlake University
Steven Dowdy, PhD
Co-Investigator

Steven Dowdy, PhD

University of California at San Diego
Allen Wang, PhD
Co-Investigator

Allen Wang, PhD

University of California at San Diego
Yuanchao Xue, PhD
Co-Investigator

Yuanchao Xue, PhD

Chinese Academy of Sciences
Peter Shaw, PhD
Project Manager

Peter Shaw, PhD

University of California at San Diego

Project Outcomes

Team Mobley anticipates that their findings will establish the foundation for developing a cell replacement therapy for the disease based on the initial proof-of-concept study. Once fully developed, the team hopes to be able to effectively reverse the disease phenotype in PD patients. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Gut-to-Brain Circuit Contributions to Parkinson-Like Phenotypes in Disease Models

Study Rationale: Although clinicians have long reported that Parkinson’s disease (PD) does not affect the brain alone, the field has only recently started to investigate gut dysfunction in experimental models of PD. Consequently, the anatomical and functional basis of gut-to-brain circuitry dysfunction in PD—such as network activity and dopamine signaling—remain poorly understood. Team Gradinaru will characterize circuit mechanisms underlying gut-to-brain disease spread and progression in the earliest appearance of Parkinson’s symptoms.

Hypothesis: Team Gradinaru’s hypothesis is that environmental and genetic factors impact the connections between neurons in the enteric nervous system (ENS), which regulates the gastrointestinal tract. This disruption may increase susceptibility to PD triggers—including aggregation of alpha-synuclein, which is toxic to cells and can trigger PD symptoms and gut inflammation. This inflammation could, in turn, augment the toxic form of alpha-synuclein that circulates to the brain and causes dysfunctions in neural circuits and motor deficits.

Study Design: Team Gradinaru will determine PD-relevant gut and brain anatomic and physiologic profiles in rodents, primates, and human cells by clarifying the anatomic pathways underlying gut-to-brain propagation of aggregated alpha-synuclein in mice, and by constructing anatomic and functional maps of macaque ENS and central nervous system PD-relevant circuits at single-cell resolution. The team also will test whether disruption of ENS circuitry mitigates gut-brain disease outcome and evaluate whether spiny mice (a rodent model that can repair damaged tissue) show protection from PD-related gut-brain degeneration.

Impact on Diagnosis: Using powerful new technologies across multiple PD-relevant model systems, this project could uncover novel circuit mechanisms that mediate symptoms, and embolden new therapeutic options to slow, halt, or perhaps reverse peripheral symptoms of PD.

Leadership
Viviana Gradinaru, BS, PhD
Coordinating Lead PI

Viviana Gradinaru, BS, PhD

California Institute of Technology
Andrew Fox, PhD
Co-Investigator

Andrew Fox, PhD

University of California at Davis
Sarkis Mazmanian, PhD
Co-Investigator

Sarkis Mazmanian, PhD

California Institute of Technology
Ashley Seifert, MSc, PhD
Co-Investigator

Ashley Seifert, MSc, PhD

University of Kentucky
David Van Valen, MD, PhD
Co-Investigator

David Van Valen, MD, PhD

California Institute of Technology
Catherine Oikonomou, PhD
Project Manager

Catherine Oikonomou, PhD

California Institute of Technology
Lin Tian, PhD
Collaborating PI

Lin Tian, PhD

Max Planck Florida Institute of Neuroscience

Project Outcomes

This project will use powerful sensor, actuator, and gene delivery technologies across multiple model systems to characterize the anatomical and functional bases of gut-brain circuitry dysfunction in PD, enabling new therapeutic options to slow, halt, or perhaps even reverse peripheral symptoms of PD. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Alpha-Synuclein Effects on Gut-Brain Circuits and Pre-Motor Symptoms in Parkinson’s Disease

Study Rationale: A hallmark of Parkinson’s disease (PD) is the development of abnormal deposits throughout the brain composed of the alpha-synuclein protein. This pathology can be found in the gut too. Increasing evidence indicates that, in some patients, alpha-synuclein pathology may begin in the gut and spread to the brain through a nerve called the vagus, which directly connects the gut to the brain. An animal model developed by Team Kaplitt’s members reproduces many of these features seen in humans to allow study of these pathways and consequences of this advancing disease as it spreads throughout the brain.

Hypothesis: Team Kaplitt hypothesizes that specific vagus nerve cells are responsible for disease spread from gut to brain, with both vagus nerve activity and other factors such as gender and menopause affecting this spread, resulting in early symptoms such as sleep disorders seen in humans before development of movement problems.

Study Design: The first two goals will use genetic manipulation to study (a) what cells may be responsible for gut-to-brain spread of abnormal alpha-synuclein, (b) how disease spread affects normal vagus functions, and (c) how different levels of vagus activity influence disease spread. Team Kaplitt will also study the consequences of this type of gut-brain spread on development of early symptoms that may occur before the movement problems, particularly sleep disorders. Given the reduced risk of PD in women prior to menopause, Team Kaplitt’s final goal is to study these same problems in a novel animal model that mimics human menopause.

Impact on Diagnosis: The gene therapy methods used to block gut-brain spread in Team Kaplitt’s studies could be applied non-invasively to patients with diagnosed presence of gut alpha-synuclein pathology to prevent disease spread. Team Kaplitt’s sleep and menopause studies will further identify opportunities for early intervention and possible hormonal approaches to limiting effects of disease spread.

Leadership
Michael Kaplitt, MD, PhD
Coordinating Lead PI

Michael Kaplitt, MD, PhD

Weill Cornell Medicine
Ted Dawson, MD, PhD
Co-Investigator

Ted Dawson, MD, PhD

Johns Hopkins Medicine
Roberta Marongiu, PhD
Co-Investigator

Roberta Marongiu, PhD

Weill Cornell Medicine
Per Svenningsson, MD, PhD
Co-Investigator

Per Svenningsson, MD, PhD

Karolinska Institute
Eileen Torres, PhD
Project Manager

Eileen Torres, PhD

Weill Cornell Medicine

Project Outcomes

Team Kaplitt anticipates that their findings will advance our collective understanding of how pathology spreads from the gut to the brain in PD, and the consequences of that pathology on early PD symptoms, with the potential for new targeted interventions to block spread and improve neuronal function. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Role of Enteroendocrine Cells in the Origin of Parkinson’s Pathology

Study Rationale: Emerging evidence supports the concept that alpha-synuclein pathology, a hallmark of Parkinson’s disease (PD), might originate in the gastrointestinal tract and spread to the brain. However, where and how pathological alpha-synuclein initially forms remains unclear. Certain gut microbiota and some environmental toxins have been associated with PD. Team Liddle recently discovered that specialized sensory cells in the gut known as enteroendocrine cells (EECs), in direct contact with gut microbiota and different environmental toxins, might transfer misfolded alpha-synuclein to the nervous system in disease.

Hypothesis: Team Liddle hypothesizes that gut microbes and environmental toxins, alone or in combination, corrupt alpha-synuclein protein expressed in EECs to misfolded pathological forms that might spread to the nervous system in a novel circuit important in PD.

Study Design: Using a unique, large, and well-characterized collection of human data, Team Liddle will identify potential triggers and alterations in gut microbiota and neuroinflammatory pathways that are associated with PD. The team will test known and experimental triggers (toxins and microorganisms) in two complementary model systems (ex vivo cultures, and in vivo in mice) to determine effects on the formation and spread of pathological alpha-synuclein within a predictable circuit of interconnected cells vulnerable to disease.

Impact on Diagnosis: These studies will (a) establish how EECs are involved in the formation of pathological alpha-synuclein at the earliest stages of disease, (b) identify gut-associated toxins and microbes that contribute to PD, (c) develop novel “humanized” pre-clinical model systems, and (d) test two particularly promising experimental therapeutic approaches in the novel model systems.

Leadership
Rodger Liddle, MD
Coordinating Lead PI

Rodger Liddle, MD

Duke University
Haydeh Payami, PhD
Co-Investigator

Haydeh Payami, PhD

University of Alabama at Birmingham
Timothy Sampson, PhD
Co-Investigator

Timothy Sampson, PhD

Emory University
Malú Tansey, PhD
Co-Investigator

Malú Tansey, PhD

University of Florida
Andrew West, PhD
Co-Investigator

Andrew West, PhD

Duke University
Senthil Gounder, PhD
Project Manager

Senthil Gounder, PhD

Duke University

Project Outcomes

Team Liddle's project will characterize specialized sensory cells of the gut, known as enteroendocrine cells, as a target for Parkinson’s disease-associated toxicant, gut microbiome, and immune interactions leading to alpha-synuclein pathology, and as a conduit to vagal, enteric, and spinal neurons. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Olfactory Circuits: Alpha-Synuclein-Rich Neurons Respond to Environmental Triggers at the Origin of Parkinson’s Disease

Study Rationale: To slow the progression of Parkinson’s disease (PD), researchers need to know more about how it starts. It is well known now that many individuals with PD have had a significant reduction in their ability to smell years before their movement disorder starts. Here, Team Schlossmacher will study the role of a PD-linked gene, alpha-synuclein, in odor processing, and how viruses in the nose may change how alpha-synuclein is handled within the odor-signaling cells of the brain in people with PD.

Hypothesis: Team Schlossmacher hypothesizes that certain environmental triggers, including viruses, can start a chain reaction inside the nasal cavity by which a protein called alpha-synuclein begins to form insoluble clumps. This happens inside the nerve circuits that specifically process scent and then gradually spreads further inside the brain, thereby promoting Parkinson’s disease.

Study Design: Team Schlossmacher will first define the normal role of the alpha-synuclein protein in the scent-processing circuits of mice and humans. The team will then answer whether so called Lewy bodies (i.e., alpha-synuclein clumps in nerve cells) contribute to the inability to smell. Third, they will study the areas of the human brain that are responsible for smell, including using MRI imaging. Lastly, Team Schlossmacher will perform studies with nasal fluids from people with PD and introduce these (and viruses) into mice, to see if these start a reaction that cause changes in the mouse olfactory system like what is seen in humans with PD, including loss of smell and changes in the alpha-synuclein protein. The team will see if the inability to smell can progress to motor deficits.

Impact on Diagnosis: This study will delineate what causes the loss of sense of smell in PD. That will help researchers to develop new drugs to treat it and explore new ways to diagnose the disease, hopefully at a stage before the movement symptoms appear. Team Schlossmacher may also gain insights into risk factors for PD, which could lead to strategies to help people reduce their risk. The team will also create new animal models that can be used by others.

Leadership
Michael Schlossmacher, MD
Coordinating Lead PI

Michael Schlossmacher, MD

The Ottawa Hospital
Benjamin Arenkiel, PhD
Co-Investigator

Benjamin Arenkiel, PhD

Baylor College of Medicine
Brit Mollenhauer, MD
Co-Investigator

Brit Mollenhauer, MD

University Medical Center Göttingen (UMG
Maxime Rousseaux, PhD
Co-Investigator

Maxime Rousseaux, PhD

University of Ottawa
Christine Stadelmann, MD
Co-Investigator

Christine Stadelmann, MD

Universität Göttingen
Julianna Tomlinson, PhD
Project Manager

Julianna Tomlinson, PhD

Ottawa Hospital

Project Outcomes

Team Schlossmacher will determine whether environment-a-synuclein interactions and inflammation in the nasal cavity can act as triggers for disease initiation, for a-synuclein aggregate formation and associated neuronal circuit dysfunction, thus informing risk and progression of disease and biomarker development in humans. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Basal Ganglia Networks in Parkinson’s Disease

Study Rationale: People with Parkinson’s disease have long been known to display remarkable motor abilities under special circumstances, such as smooth walking with certain visual or auditory cues. This phenomenon is called paradoxical kinesia. In addition, placebos can be surprisingly effective in treating the motor signs of the disease. Team Strick hypothesizes that a specific neuroanatomical substrate supports paradoxical kinesia and the placebo effect. The team plans to define this substrate and investigate its functional organization.

Hypothesis: Team Strick hypothesizes that a specific neural circuit supports paradoxical kinesia and the placebo effect.

Study Design: Team Strick will use cutting-edge techniques to reveal the two distinct brain circuits that enable the basal ganglia to influence the control of voluntary movement in primates. Next, the team will record the electrical and chemical activity of basal ganglia neurons in the best animal model of Parkinson’s disease. In addition, Team Strick will determine the molecular signatures of basal ganglia neurons that are affected by the disease and those that are left intact. Finally, the team will image neural activity in human subjects affected by the disease to determine the full range of strategies that could be used to improve basal ganglia function.

Impact on Diagnosis: Team Strick’s results could re-shape paradigms for therapeutic development and attempts to influence disease progression. Importantly, the team’s results have the potential to use basal ganglia circuits that are untouched by the disease to promote recovery of more normal motor function..

Leadership
Peter Strick, PhD
Coordinating Lead PI

Peter Strick, PhD

University of Pittsburgh
Scott Grafton, MD
Co-Investigator

Scott Grafton, MD

University of California at Santa Barbara
Helen Schwerdt, PhD
Co-Investigator

Helen Schwerdt, PhD

University of Pittsburgh
William Stauffer, PhD
Co-Investigator

William Stauffer, PhD

University of Pittsburgh
Robert Turner, PhD
Co-Investigator

Robert Turner, PhD

University of Pittsburgh
Andreea Bostan, PhD
Project Manager

Andreea Bostan, PhD

University of Pittsburgh

Project Outcomes

Team Strick's project will perform a multidisciplinary characterization of the networks that link the basal ganglia with the cortical motor areas. The information from the team's studies could lead to new therapeutic targets to delay disease progression and new approaches to ameliorate the motor dysfunction that is so disabling for PD patients. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Distributed Circuit Dysfunction Underlying Motor and Sleep Deficits in a Progressive Model of Parkinson’s Disease

Study Rationale: Parkinson’s disease (PD) begins decades before it compromises the ability to move about in the world and sleep through the night. Understanding how the dysfunction of brain circuits begins and then evolves to cause difficulty in moving and sleeping will allow researchers to diagnose PD earlier—increasing the chances of halting disease progression—and to better treat the disease once it appears.

Hypothesis: The progressive damage to dopamine-releasing neurons results in staged disruption of neural circuits in larger and larger parts of the brain, ultimately leading to both motor and sleep deficits characteristic of PD.

Study Design: Team Surmeier’s plan is to study a new genetically engineered mouse model that manifests a progressive, levodopa-responsive parkinsonism. Importantly, this mouse faithfully reproduces the human staging of pathology in key brain circuits. Using the most advanced methods available for studying and manipulating genetically defined brain circuits, the causal linkage between circuit dysfunction and motor and sleep behavior will be determined. 

Impact on Diagnosis: A better understanding of how the circuit dysfunction underlying PD is staged should allow earlier diagnosis—enhancing the potential benefit of disease-modifying therapies—and better treatment strategies for later-stage patients.

Leadership
D. James Surmeier, PhD
Coordinating Lead PI

D. James Surmeier, PhD

Northwestern University
Silvia Arber, PhD
Co-Investigator

Silvia Arber, PhD

University of Basel
Rui Costa, PhD, DVM
Co-Investigator

Rui Costa, PhD, DVM

Allen Institute
Yang Dan, PhD
Co-Investigator

Yang Dan, PhD

University of California at Berkeley
Ann Kennedy, PhD
Co-Investigator

Ann Kennedy, PhD

Northwestern University
Chrissy Weber-Schmidt
Project Manager

Chrissy Weber-Schmidt

Allen Institute

Project Outcomes

Team Surmeier's project will provide fundamental insight into the relationship between progressive dopamine depletion and distributed circuit dysfunction underlying motor and sleep symptoms of Parkinson’s disease. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Activity and Connectivity Drive Neuronal Vulnerability and Disease Progression in Parkinson’s Disease

Study Rationale: Specific brain circuits that are highly melanized (build-up of the dark pigment neuromelanin) with age are primarily affected, particularly early, in Parkinson’s disease (PD). Models incorporating this aspect of PD have only been developed recently and show that increased neuromelanin production causes neurodegenerative changes consistent with Parkinson’s. The regulators of cellular neuromelanin metabolism have not been determined, the effect of neuromelanin on normal activity in these pathways has not been defined, the potential for neuromelanin aggregates to increase alpha-synuclein accumulation has not been evaluated, and the impact of extracellular neuromelanin on detrimental inflammatory processes has not been assessed.

Hypothesis: Activity in melanized brain circuits is a dominant factor in the initiation of PD and sustains its progression by seeding pathology in connected regions and providing the stimulus for chronic inflammation. Manipulating neuromelanin production and/or brain circuit activity can ameliorate these deficits. 

Study Design: Parallel experiments will be performed in mice and non-human primates in which neuromelanin production has been induced for comparison with neuromelanin-producing neurons in people with prodromal and early Parkinson’s. To test whether activity in melanized brain circuits is a dominant factor in the initiation of PD, spatiotemporal activity mapping, imaging, and other techniques will be used, and manipulating neuromelanin production and/or brain circuit activity will be assessed as potential treatments. To determine if neurons spread PD pathology through their connectivity, seeding experiments will be performed and impacts on behavior and neurodegeneration assessed. To determine how non-neuronal mechanisms are involved in disease progression, high-resolution microscopy and cell-specific details of changes in extracellular spaces and infiltration of non-neuronal cells into the brain will be assessed.

Impact on Diagnosis: Diagnosis of neuromelanin changes in the brain are already being assessed for their diagnostic potential, but this study will determine their focus and rate of change with respect to neural activity and clinical features. Team Vila will also identify if reducing neuromelanin levels stabilizes pathology and restores brain activity.

Leadership
Miquel Vila, MD, PhD
Coordinating Lead PI

Miquel Vila, MD, PhD

Vall d'Hebron Research Institute (VHIR)
Glenda Halliday, PhD
Co-Investigator

Glenda Halliday, PhD

University of Sydney
Nicola Mercuri, MD
Co-Investigator

Nicola Mercuri, MD

Tor Vergata University
Jose Obeso, MD, PhD
Co-Investigator

Jose Obeso, MD, PhD

Network Center for Biomedical Research in Neurodegenerative Diseases
Matthias Prigge, PhD
Co-Investigator

Matthias Prigge, PhD

Leibniz Institute for Neurobiology
Javier Hoyo, PhD
Project Manager

Javier Hoyo, PhD

Vall d'Hebron Research Institute (VHIR)

Project Outcomes

The project will unravel molecular mechanisms linking brain circuit activity to PD vulnerability, identify brain circuits through which PD pathology spreads across the brain and periphery, establish non-neuronal mechanisms of PD progression and determine whether modulation of neuromelanin levels and/or brain circuit activity can restore PD circuit dysfunction & pathology. View Team Outcomes.

Expand to Read More +Collapse to Read Less –
Circuitry and Brain-Body Interactions | 2021

Cortical Pathophysiology of Parkinsonism

Study Rationale: The outer mantle of the brain, the cerebral cortex, plays a significant role in selecting and controlling movements. Changes in the activity of cortical neurons are key to disorders of movement, especially Parkinson’s disease (PD). It is unknown, however, which specific cell types are involved and how their activity changes during the course of the disease. In these experiments, Team Wichmann will use new technologies to study large groups of specific types of cortical neurons (for example, those that send fibers to the spinal cord) and explore how their activity and morphology change in animal models of chronic PD.

Hypothesis: Team Wichmann’s hypothesis is that groups of cortical neurons that send fibers to the spinal cord—unlike those that send projections to the striatum—start to show abnormal activity and undergo morphological changes in connections that provide inputs to them when parkinsonism develops.

Study Design: Team Wichmann will measure the anatomical and functional characteristics of neurons in the motor cortex in animal models of slowly progressive PD. Optical imaging methods as well as electrophysiologic recordings will allow the team to measure the activity patterns of large groups of individual cortical neurons, while parallel anatomical studies will identify the reshaping of connections to different families of cortical neurons before and during the development of parkinsonism. Computational analysis will allow Team Wichmann to put the findings together in computer simulations that will help them to understand the cortical circuit abnormalities that contribute to PD.

Impact on Diagnosis: A better understanding of how movement problems in PD develop is key to developing more effective methods to control them. Characterizing the abnormalities in specific families of cortical neurons may allow researchers to develop new therapies that target the affected circuits through deep brain stimulation, pharmacologic, or genetic methods.

Leadership
Thomas Wichmann, MD
Coordinating Lead PI

Thomas Wichmann, MD

Emory University
Hong-Yuan Chu, PhD
Co-Investigator

Hong-Yuan Chu, PhD

Van Andel Research Institute
Adriana Galvan, PhD
Co-Investigator

Adriana Galvan, PhD

Emory University
Yoland Smith, PhD
Co-Investigator

Yoland Smith, PhD

Emory University
Johnson Agniswamy, PhD
Project Manager

Johnson Agniswamy, PhD

Emory University

Project Outcomes

The activity and anatomy of neurons in the brain’s outer mantle, the cortex, are abnormal in Parkinson’s disease. Team Wichmann's studies will help researchers to understand which specific cells or connections are involved in parkinsonism. This knowledge may allow researchers to therapeutically target these circuits through stimulation, pharmacologic, or genetic methods. View Team Outcomes.

 

The ASAP Collaborative Research Network spans beyond circuitry interactions. Check out the functional genomics and neuro-immune themes for a complete look at the work our diverse group of grantees is doing to tackle key knowledge gaps in Parkinson’s disease research.