Recognized Excellence of Allergy and Immunology Division Trainees, Fellows and Associates

The Division of Allergy and Immunology is proud of the excellence of its undergraduate and graduate trainees, postdoctoral research and clinical fellows and associates. Several received recognition for their achievements throughout the year:

  • Amy Eapen, MD, a clinical fellow graduating from the Allergy / Immunology Fellowship Program, contributed one book chapter and one manuscript to the literature under the mentorship of Leah Kottyan, PhD, and received a 2019 AAAAI Travel Award. She accepted a position as an assistant professor at Wayne State University.
  • William Lavery, MD, PhD, a clinical fellow graduating from the Allergy / Immunology Fellowship Program, contributed one book chapter and four manuscripts to the literature under the mentorship of Artem Barski, PhD, and Andrew Lindsley, MD, PhD.
  • Vanessa Gadoury-Levesque, MD, a clinical fellow graduating from the Allergy / Immunology Fellowship Program, received the Best Poster Award at the 2019 Canadian Society of Allergy and Clinical Immunology (CSACI) Annual Scientific Meeting and contributed one publication to the literature under the mentorship of Risma.
  • Steven Proper, DO, PhD, a clinical fellow graduating from the Allergy / Immunology Fellowship Program, contributed one manuscript to the literature under the mentorship of Gurjit Khurana Hershey, MD, PhD, and presented research at the 2020 AAAAI and 2020 Annual Thomas F. Boat Lectures.
  • Ian Slack, MD, a first-year clinical fellow of the Allergy / Immunology Fellowship Program, contributed one manuscript to the literature under the mentorship of J. Pablo Abonia, MD, and received a 2019 ACAAI Travel Scholarship Award for his poster presentation “Recurrent Volvulus as a Herald Sign for Idiopathic Systemic Capillary Leak Syndrome.”
  • Katharine Guarnieri, MD, a first-year clinical fellow of the Allergy / Immunology Fellowship Program, received a 2019 ACAAI Tavel Scholarship Award for her oral presentation “Erythema Multiforme-Like Rashes: Are They Truly Antibiotic-Associated”.
  • Nurit Azouz, PhD, a research fellow in the Rothenberg CURED Lab, won an International Eosinophil Society Travel Grant Award to attend and present at the International Eosinophil Society 2019 Biennial Symposium. Nurit presented her abstract “Functional role of kallikrein 5 and proteinase activated receptor 2 in eosinophilic esophagitis".
  • Tetsuo Shoda, MD, PhD, a research fellow in the Rothenberg CURED Lab, won an International Eosinophil Society Travel Grant Award to attend and present at the International Eosinophil Society 2019 Biennial Symposium. Tetsuo presented his poster “Molecular, endoscopic, histological and circulating biomarker-based diagnosis of eosinophilic gastritis: cross sectional multi-site study".
  • A special thank you to Stephanie L. Ward, MD, for her continued leadership in educational mentorship of medical students with the University of Cincinnati and to Risma for her continued leadership in educational mentorship of Allergy / Immunology fellows and as leader of the Allergy / Immunology Fellowship Program.

Food Allergy Program

The Food Allergy Program, led by Assa’ad improves the lives of patients with food allergies and their family members by providing expert care, innovative treatments and cutting-edge research. In addition to providing patient services, engaging in clinical trials and leading the field in improving oral immunotherapy protocols, the Food Allergy Program garnered two key research awards and published seven journal articles in fiscal year 2020, including research investigating oral immunotherapy for multiple foods (Eapen et al.); the effects of age, sex, race/ethnicity, health insurance and food-specific serum immunoglobulin E on outcomes of oral food challenges (Dang et al.); prevention of food allergy (Devonshire et al.); non-immunoglobulin E-mediated food allergy during infancy (Devonshire et al.); and the allergenicity of black walnut tree syrup (Lierl et al.)

Food Allergy Program Garners Two Key Awards

Cincinnati Children’s Food Allergy Program received two awards that will ultimately benefit patients with food allergy by adding to its program and creating new infrastructure to promote additional research. The program, led by Assa’ad, is a Food Allergy Research & Education (FARE) Clinical Research Center of Distinction. This means that Cincinnati Children’s now will be part of a national network carrying out multicenter investigations and other clinical research, training the next generation of food allergy investigators and delivering optimal clinical care and leadership in the community to patients with food allergy. In addition, the Food Allergy Program received a $2.5 million grant from FARE to be its Clinical Network Centers’ (FCN) Biobank and Biomarker Discover Center (BBDC). The BBDC will be in charge of the development of critical biorepository infrastructure that is necessary to carry out the FCN mission and will serve as a tactical hub for biospecimens and a key participant in the design and implementation of food allergy related biomarker and translational research. The new biobank is a collaborative effort with the Division of Biostatistics and Epidemiology and the Discover Together Biobank and will be a valuable resource to change the outcome together in food allergy research into the future.

FDA Approves New Drug to Help Children with Peanut Allergy

If your child has a peanut allergy, life-changing help could be on the way. In February 2020, the Food and Drug Administration (FDA) approved a new drug tested at Cincinnati Children's. Amal H. Assa’ad, MD, director of the Food Allergy Program, said that it is not a cure but can save lives. The FDA-approved new drug, Palforzia, may ease the severe and sometimes deadly reactions from eating or even being near peanuts. Assa'ad had been testing Palforzia with pediatric patients at Cincinnati Children's since 2014. Children with peanut allergies received extremely small amounts of peanut protein, and then doctors increased the dosage over time. By 12 visits, patients reached a dosage amount that they could maintain, about a peanut to a peanut in a half. Once reached, daily maintenance of that dose must occur for the rest of their lives. The therapy is currently limited to peanut allergies and only intended for children ages 4-17, but application of this same process in the future may help with other food allergies. Learn more about the Assa'ad Lab.

Predicting Risk of Peanut Allergy in Infants

Ashley L. Devonshire, MD, MPH, received a KL2 Research Scholar Award from the Center for Clinical and Translational Science and Training (CCTST) at the University of Cincinnati to investigate the molecular signature from CD4+ T cells in infants with clinically reactive peanut allergy versus infants with peanut tolerance. Resulting data will aid in developing tools to predict risk of peanut allergy in infants and to expand our knowledge of food allergy in the understudied infant population. Learn more about the Devonshire Lab.

New Faculty: Yasmin Hassoun, MD

The division welcomes Hassoun as a faculty member. Hassoun is an internal medicine physician who specializes in food allergy and allergic rhinitis. Medicine is a lifelong interest for Hassoun, starting with her first exposure to the field of allergy and immunology during her internal medicine training. During that training, she saw firsthand the impact that a diagnosis of allergy has on children and their families and how working with the physician can alleviate some concerns, clear up misconceptions and offer treatment plans. Hassoun believes that the patient-physician relationship is very important and that empowering the patient and family to take control of their diagnosis, speak up and ask questions is essential. Establishment of the best patient care is through an effort of both the patients, their families and the physician.

Bringing Genomics and Epigenetics to Bear on Allergic Mechanisms

Barski, co-developer of ChIP-seq, garnered three awards in fiscal year 2020 to fund research using or developing genomics and epigenetics techniques to investigate allergic mechanisms. GAP funding from Cincinnati Children’s supports “An experimentally refined, dynamic gene regulatory network model of T-cell memory” in collaboration with Emily R. Miraldi, PhD, of the Divisions of Immunobiology and of Biomedical Informatics, and pilot funding by the Digestive Health Center supported “Following allergen-specific T cells in food allergy”. Notably, the National Institute of General Medical Sciences (NIGMS) / National Institutes of Health (NIH) R21 funding supporting “Death-Seq, a Method for Genome-wide Identification of Functional Silencer Elements” will advance methodology applicable across disciplines for future biological research. Learn more about the Barski Lab.

Transition and Adult Primary Immune Deficiency Program

Cincinnati Children’s is a worldwide leader in the diagnosis and treatment of primary immune deficiency diseases. More immune deficiency diseases are now recognized in adults, and children with primary immune deficiencies eventually need to transition to adult care providers. To fill these important care needs, Erinn S. Kellner, MD, founded the Transition and Adult Primary Immune Deficiency Program, which focuses on the complex needs of adults and adolescents with primary immune deficiencies and basic, translational and clinical research to understand the mechanisms of different immune deficiencies and how they may change over time. The Transition and Adult Primary Immune Deficiency Program collaborates with adult subspecialty services at the University of Cincinnati and the Cincinnati Children’s Diagnostic Immunology Laboratory and Immune Deficiency and Histiocytosis Program. Learn more about the Kellner Lab.

Drug Allergy Program

Kimberly A. Risma, MD, PhD, formalized and expanded inpatient and outpatient services to address unmet needs for children with drug allergies by founding the Drug Allergy Program at Cincinnati Children’s. The Drug Allergy Program diagnoses and treats drug allergies in children in the Greater Cincinnati area and in patients referred nationally. An important aspect of the program is “de-labeling” drug allergies by testing children for a clinical reaction to the drug in a controlled setting. For instance, recent studies show that 95% of children labeled as having an “allergy” to amoxicillin do not have a clinical reaction when re-exposed to it through testing. Delabeling drug allergies prevents use of alternative drugs that may cost more, have more side effects, or contribute to increased antibiotic resistance in our communities.

Mechanisms and Metrics for Esophageal Eosinophilia

Wen received two funding awards in fiscal year 2020 directly related to his innovative findings in tissue-resident lymphocytes. The National Institute of Allergy and Infectious Diseases (NIAID) / National Institutes of Health (NIH) R01 will support his translational research investigating the roles of the FFAR3-SCFA axis in Th2 cytokine production by tissue lymphocytes in eosinophilic esophagitis (EoE), and the National Institute on Aging (NIA) funded a complementary supplement for fiscal year 2021 exploring the role of this FFAR3 axis in the context of aging. The Center of Pediatric Genomics (CPG) Innovation Award will support a corresponding project assessing whether oral squamous epithelial cell gene expression profiles will reliably and consistently predict human esophageal tissue eosinophilia with high precision, which would represent an advance in non-invasive disease activity monitoring for EoE. Learn more about the Wen Lab.

Schwartz Lab Research

The Schwartz Lab conducts translational research focusing on the roles of innate immune cells in the development, progression and resolution of allergic disease in children. In fiscal year 2020, Justin Schwartz, MD, PhD, received two funding awards. He is the principal investigator of a translational research project supported by the National Institute of Allergy and Infectious Diseases (NIAID) / National Institutes of Health (NIH) Inner City Asthma Consortium (ICAC). The ICAC studies monitoring and immune-based treatments with the potential to help inner city and disadvantaged children with asthma. Schwartz also conducts an industry-sponsored translational study with Knopp Biosciences.

Industry Clinical Trials

The Division of Allergy and Immunology at Cincinnati Children’s believes that research is the hope for future clinical advances in care and treatment. In fiscal year 2020, there were six newly initiated clinical trials in the division. Assa’ad leads two investigating atopic dermatitis: 1) “Evaluation of Biomarkers of Atopic Dermatitis in Pediatric Patients Whose Disease is Not Adequately Controlled with Topical Prescription Therapies Or When Those Therapies Are Note Medically Advisable” with Sanofi (ClinicalTrials.gov Identifier: NCT03849716); and 2) “An Open-label Multiple-Dose Study to Evaluate the Pharmacokinetics, Safety, and Tolerability of Upadacitinib in Pediatric Subjects with Severe Atopic Dermatitis" with AbbVie (ClinicalTrials.gov Identifier: NCT03646604). Rothenberg leads two investigating eosinophilic esophagitis (EoE): 1) “A Multicenter, Randomized, Double-Blind, Parallel-Group, Placebo-Controlled Study to Investigate the Use of Benralizumab for Eosinophilic Esophagitis (MESSINA)” with AstraZeneca (EudraCT number: 2019-002871-32); and 2) “A Phase 3, Randomized, 3-Part Study to Investigate The Efficacy And Safety of Dupilumab in Adult and Adolescent Patients with Eosinophilic Esophagitis” with Regeneron Pharmaceuticals (ClinicalTrials.gov Identifier: NCT03633617). Sandy Durrani, MD, leads two investigating EoE and eosinophilic gastritis: 1) "A Phase 2/3, Multicenter, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Efficacy and Safety of AK002 in Adult and Adolescent Patients with Active Eosinophilic Esophagitis" with Allakos, Inc. (ClinicalTrials.gov Identifier: NCT04322708); and 2) "A Phase 3, Multicenter, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Efficacy and Safety of AK002 in Patients with Moderately to Severely Active Eosinophilic Gastritis and/or Eosinophilic Gastroenteritis Who Have an Inadequate Response with, Lost Response to, or Were Intolerant to Standard Therapies" with Allakos, Inc. (ClinicalTrials.gov Identifier:NCT04322604).

FDA-Approved Drug Reverses Inflammation in Animal Model of Eosinophilic Esophagitis

Scientists at Cincinnati Children’s discovered an imbalance of specific proteins, proteases and protease inhibitors in cells lining the esophagus may cause inflammation and tissue damage in people with eosinophilic esophagitis (EoE). People with EoE experience difficult or painful swallowing, vomiting and nutritional problems, and EoE is histologically hallmarked by an accumulation in the esophagus of immune cells called eosinophils. In people with active EoE, biopsies of esophageal tissues show a near-complete lack of the protease inhibitor, SPINK7, which normally damps down inflammation and helps preserve tissue structure. However, Nurit Azouz, PhD, and Rothenberg led a study published in Science Translational Medicine reporting that the biologic drug alpha-1 anti-trypsin (A1AT) reversed damaging inflammation in an animal model of EoE. The finding suggests that further investigation warrants determination on whether the therapeutic may also benefit people with EoE. Current treatments for EoE include often-restrictive diets to avoid trigger foods, corticosteroids to relieve inflammatory symptoms and sometimes surgical procedures to expand the esophagus. These findings highlight esophageal allergy as a protease-mediated disease and that disarming of proteases by delivering protease inhibitors has the potential to be therapeutic in allergic disease. Read this blog post to learn more.

High-Tech Tissue Study Reveals Which Cells Drive a Painful Food Allergy

The lifelong allergic disease eosinophilic esophagitis (EoE) occurs when too many eosinophils (a type of white blood cell) cause local inflammation that makes swallowing and eating painful. EoE can be so serious that some people depend upon special liquid formulas to eat. An eight-year hunt for the cells that drive EoE identified a disease-causing T cell subtype along with a potential new way to treat the disease while also raising questions about a dietary supplement often taken to reduce bowel inflammation. In this Journal of Clinical Investigation study, Ting Wen, MD, PhD, and Rothenberg developed and perfected a two-hour process that isolates and analyzes individual living cells from human tissue samples. This is a substantial advance for the field, as prior work focused on blood cells, but not the specific memory immune cells in tissue that directly respond to food allergens and induce eosinophilic inflammation. Wen and Rothenberg uncovered the unknown properties of these cells, including these key findings:

  • The discovery of eight types of tissue-resident T cells in the esophageal tissue is more than previously expected, and more than previously studied.
  • Two cell types (T7 and T8) show up in higher numbers in the diseased tissue. The study finds that the latter (T8) cell type responds to allergens by producing massive amounts of type 2 cytokines, an inflammation-causing substance.
  • One of these cell types (T8) is nearly non-existent in healthy tissue as assessed by known and novel surface markers, making it especially worth targeting for treatment. Inhibiting these cells is now a central goal of further research in the Wen Lab.
  • One of the overactive genes that helps trigger the inflammation response—a gene called FFAR3—also encodes a receptor for a dietary supplement called butyrate, which is commonly seen in normal and EoE diets.

Beyond EoE, the process used in this study is applicable in other, more-common conditions to find small populations of cellular culprits hiding within tissues. The study co-authors say that the process is possibly useful in studying asthma, atopic dermatitis, allergic rhinitis and other diseases associated with type 2 cytokines. Read more in this blog post.

New Tests May Help Diagnose Rare Subset of Eosinophilic Disorders

As a group, eosinophilic gastrointestinal disorders (EGIDs) are rare and often hard to diagnose. The most common form—eosinophilic esophagitis (EoE)—affects an estimated 158,000 of more than 320 million people in the United States. Other forms, such as eosinophilic gastritis (EG), are much less common, which makes them even more difficult to accurately diagnose. A study in the Journal of Allergy and Clinical Immunology led by Shoda and Rothenberg at Cincinnati Children’s introduces new tissue and blood testing platforms that may make it easier for specialists to pinpoint EG. Patients with EG often experience a long journey to correct diagnosis and effective treatment. For years, there are not any standardized diagnostic criteria, and there is little known about the causes of EG. These difficulties are largely due to EG being very rare and most patients with EG also having other EGIDs. With fewer individuals to treat and study, there are fewer opportunities to learn more about this condition and draw conclusions. The new study builds upon earlier work and the histologic characterization of EG in 2014 (Caldwell et al. JACI 2014). Shoda and colleagues examined gastric biopsy and blood samples from 185 individuals from nine sites of the Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR) to develop improved diagnostic platforms for EG. The biopsy and blood samples allowed the research team to develop a molecular profile that showed robust associations with histologic and endoscopic features. Plasma eotaxin-3 levels strongly associate with gastric CCL26 expression, making this relationship the strongest single tissue and circulating disease biomarker. These new tissue- and blood-based platforms for assessing EG are possible by collaboration among CEGIR members, including divisional colleague Abonia. Before launching a prospective clinical trial to validate these platforms, explore their feasibility in various clinical settings and ultimately optimize the platforms for disease stratification requires further work. Read this blog post to learn more.

Cincinnati Children's Receives NIH Grant Renewal to Continue Leading Multicenter Consortium on Eosinophilic Disorders

Cincinnati Children’s received a five-year, $7.57 million grant renewal from the National Institutes of Health (NIH) to continue leading a consortium of organizations (CEGIR) from around the country to conduct clinical research into eosinophilic gastrointestinal disorders. The belief is that allergic hypersensitivity to certain foods and an over-accumulation in the gastrointestinal tract of white blood cells, called eosinophils, trigger chronic inflammatory conditions . Eosinophilic disorders can cause a variety of gastrointestinal complaints, including reflux-like symptoms, vomiting, difficulty swallowing, tissue scarring, fibrosis, the formation of strictures, diarrhea, abdominal pain and failure to grow in childhood. In the previous grant, researchers studied inflammation in the esophagus, stomach and colon. The new grant will focus on these three conditions in addition to eosinophilic gastroenteritis. Led by Rothenberg, and Glenn Furuta, MD, CEGIR will continue to further research and develop clinical expertise, train clinical investigators, pilot clinical research projects and provide access to information related to eosinophilic disorders for basic and clinical researchers, physicians, patients and the lay public. CEGIR receives funding from the Rare Diseases Clinical Research Network (RDCRN) and comprises researchers from 15 institutions and three major patient advocacy groups, including the Campaign Urging Research for Eosinophilic Disease (CURED), American Partnership for Eosinophilic Disorders (APFED), and the Eosinophilic Family Coalition (EFC). Read this news release to learn more. Read this blog post to learn more.

Fifth CURED Research Conference and Patient Education Program

Cincinnati Children’s hosted the 5th CURED Research Conference and Patient Education Program, an international meeting focusing on the emerging field of eosinophilic gastrointestinal disorders (EGID), including eosinophilic esophagitis (EoE). The conference took place Nov. 7-9, 2019, and addressed clinical, pathogenic, genetic, dietary and therapeutic features of EGID. The program featured renowned international experts speaking over three days, with the first two days focused on EGID research and the third day on patient and family education. The conference accepted over 40 abstracts for poster presentations, with many late-breaking discoveries presented as oral presentations. The Campaign Urging Research for Eosinophilic Disease (CURED) Foundation in collaboration with the Rothenberg CURED Lab, Cincinnati Center for Eosinophilic Disorders (CCED), and the Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR), with funding from the National Institutes of Health (NIH), held the event. This is the fifth such meeting and attendance reached nearly 300 individuals from 20 states in the USA, seven countries and four continents. Read this blog post for more information about this conference.

EGIDExpress Tool Helps Accelerate EoE Research

Adina Ballaban, BS, of the Rothenberg CURED Lab created a platform dubbed EGIDExpress for the rapidity and access that it gives to the datasets generated for research of eosinophilic gastrointestinal disorders (EGIDs). Like an expressway or express train, it acts as a vital infrastructure in directly and quickly reaching destinations. EGIDExpress moves eosinophilic disease research forward by serving as a platform to spur ideas and accelerate generation of hypotheses related to eosinophilic disease, with the goal of ultimately advancing research. Adina and the Rothenberg CURED Lab worked with the Division of Biomedical Informatics to make this vision a reality, launching EGIDExpress to the public in November 2019. However, it is not a static resource, with added datasets as they become available. EGIDExpress is for data sharing and offers not only researchers, but also the public the opportunity to view published datasets generated by the Rothenberg CURED Lab on demand. Through this web-app, data can be easily accessed and readily visualized on a per gene basis or conveniently downloaded in bulk. As molecular and personalized medicine continue to advance for EGIDs, public availability to knowledge via EGIDExpress is allowing people to explore the personal relevance of recent findings. Read this blog post to learn more.

Typing Eosinophilic Esophagitis

Eosinophilic esophagitis (EoE) is a chronic inflammatory disease largely triggered by immune hypersensitivity to food and characterized by eosinophils in esophageal tissue. Emerging data substantiate the importance of type 2 cytokines in disease pathogenesis, yet systematic analyzation of the variability of type 2 immunity needs to occur. Julia Dunn, PhD, and Rothenberg molecularly profiled esophageal biopsies from 312 patients with EoE from 10 sites associated with the Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR). In a study published in the Journal of Allergy and Clinical Immunology, the authors identified five groups of patients with distinct type 2 cytokine expression despite similar eosinophil levels. Synthesizing these data with recently characterized EoE endotypes, the authors propose a disease mechanism that transitions from IL5-low/normal to IL5-high/inflammatory and finally to IL5-intermediate/fibrostenotic processes. These studies identified unique patterns of inflammatory gene expression and lay groundwork for future studies to predict EoE responsiveness to treatments, including those targeting type 2 immunity in order to improve outcomes.

Binational Collaborative Eosinophilic Esophagitis Research

Eosinophilic esophagitis (EoE) is an emerging, chronic, allergic disease with dramatic and continuous increases in prevalence in nearly all regions of the world, including the US, Europe and Israel. EoE has one of the lowest qualities of life, likely because of the restricted diets, chronic pain, relapsing nature and need for recurrent endoscopies. The Binational Science Foundation (BSF)’s Regular Grants Program awarded Ariel Munitz, PhD, of Tel Aviv University, Israel, and Marc E. Rothenberg, MD, PhD, of Cincinnati Children’s grant funding to investigate the role of the type 2 IL-4 receptor in EoE, define the relative contribution of IL-4 and IL-13 to EoE pathogenesis via this receptor chain, and assess whether pharmacologic targeting of the type 2 IL-4 receptor via neutralization of IL-13 receptor α1 can alleviate EoE. This collaborative research project is an exciting opportunity to better understand disease pathogenesis and develop more effective, mechanism-based precision therapy (BSF Award and Abstract).

Director Becomes President Elect of International Eosinophil Society

Rothenberg became president elect of the International Eosinophil Society (IES). Rothenberg is the principal investigator for the Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR) of the Rare Diseases Clinical Research Network (RDCRN) and director of the Cincinnati Center for Eosinophilic Disorders at Cincinnati Children’s. In 2019, he received the Paul Ehrlich Lectureship Award, the highest honor bestowed by the IES to an individual who makes major contributions to the advancement of our understanding of the eosinophil. Rothenberg’s research focuses on molecular analysis of allergic inflammation, primarily on the molecular pathogenesis of eosinophilic gastrointestinal disorders (EGIDs). His Rothenberg CURED Lab takes a multi-disciplinary approach including the development of preclinical murine models: genetics, genomics, molecular immunology, and biochemistry. The laboratory has several broad research areas and objectives, including the epigenetic and genetic basis of allergic disease and responses, immunopathogenesis of allergic inflammation, impaired barrier function, pathogenesis of EGIDs, precision and predictive medicine, and proteases and protease inhibitors in inflammation. Nationally, Rothenberg leads CEGIR, which dedicates itself to improving the lives of individuals with EGIDs through innovative research, clinical expertise and education via collaborations between scientists, health care providers, patients and professional organizations.

Zimmermann Lab Research

Nives Zimmermann, MD, PhD, is a pathologist with a research focus on pediatric pathology, especially related to asthma, eosinophils, lung inflammation and diseases that affect blood cells. Her research concentrates on eosinophilic diseases, specifically why some patients get them and others do not. Eosinophils can be present in some tissues with no ill effects, whereas at other times or in other tissues they cause serious damage. The Zimmermann Lab investigates mechanisms that cause this damage. Zimmermann recently completed a residency in Clinical and Anatomic Pathology at the University of Cincinnati; while pursuing this pathology specialty training, Zimmermann’s dedication received recognition with the Pathology Department of the University of Cincinnati Disciplined Developer Award and the Resident of the Year Award.

Lindsley Lab Research

Lindsley received research support through a Center for Pediatric Genomics pilot grant to unravel the mechanisms of epigenetic neurocristopathy in Kabuki syndrome. The major goal of the projects is to determine the cellular and epigenetic mechanism by which KMT2D mutations drive craniofacial defects. The Lindsley Lab aims at understanding how genetic and epigenetic mechanisms alter immune responses in immune-mediated diseases, focusing on: 1) the role of chromatin regulation in B and T cell differentiation / function (with a special focus on Kabuki syndrome); and 2) the effects of genetic risk variants on promote pediatric asthma. Lindsley serves as a Medical Advisory Board member for the Kabuki Syndrome Foundation and All Things Kabuki, a patient advocacy group.