Clinicians

Eva Morava MD PhD

Professor of Medical Genetics; Consultant

Division of Medical Genetics and Genomics Sciences, Mount Sinai

Eva Morava-Kozicz, M.D., Ph.D., conducts translational research in mitochondrial disorders and congenital disorders of glycosylation (CDGs). Dr. Morava is actively involved in developing dietary therapies in genetic disorders and is the principal investigator of a multicenter study on the natural history of CDG.

Focus areas

  • Diagnostics therapy and follow-up of mitochondrial disease. Dr. Morava’s modified scoring system has been used for more than 10 years to diagnose mitochondrial disease. It is now being adapted for the next-generation sequencing era to discover novel types of mitochondrial disorders, including the novel group of phospholipid synthesis defects.
  • Discovering new congenital disorders of glycosylation. Using next-generation sequencing, Dr. Morava played a crucial role in the definition of ATP6V0A2-CDG, SRD5A3-CDG, SLC35A1-CDG, ATP6V1A-CDG, ATP6V1E1-CDG and PGM1-CDG. She was also involved in the discovery of DPM2-CDG, DPM1-CDG, COG7-CDG, MAN1B1-CDG, ATP6VAP1-CDG, CCDC115-CDG and TMEM199-CDG, and is currently involved in unsolved disease discovery.
  • Developing therapies for CDGs. Dr. Morava is involved in evaluating therapies in CDGs, including transplantation and dietary therapies such as D-galactose therapy. She is investigating the D-galactose working mechanism and is involved in clinical trials of many types of CDGs. Dr. Morava is working in close collaboration with the patient associations United Mitochondrial Disease Foundation (UMDF) and CDG CARE.
  • Defining the new syndrome group of metabolic cutis laxa. Previously called ARCL type 2A, metabolic cutis laxa is a connective tissue disorder that causes premature aging. Dr. Morava first described the glycosylation abnormalities in ARCL type 2A, and defined the characteristic phenotype of the new disease, metabolic cutis laxa. Since then, several new inborn errors have been found with different metabolic abnormalities.

Links and key research papers

Andrew C. Edmondson, MD, PhD

Attending Physician

Metabolic Disease Program and the Division of Human Genetics at Children’s Hospital of Philadelphia.

Research Interests: We use model systems (primary cells, iPSCs, mouse) of rare genetic neurodevelopmental disorders that disrupt glycosylation (known as Congenital Disorders of Glycosylation, or CDG) and glycoproteomics to study glycosylation in the brain.

Research Details: Glycosylation is the enzyme-mediated process by which a carbohydrate (or “glycan”) is covalently attached to a target macromolecule (typically a protein or lipid). As the most abundant post-translational modification, glycosylation generates immense biological variability and mediates fundamental biological processes. The biological significance of glycosylation is emphasized by the Congenital Disorders of Glycosylation (CDG), a group of ~170 rare genetic diseases that disrupt glycosylation. CDG patients exhibit multiorgan dysfunction, including neurological deficits such as epilepsy and neurodevelopmental abnormalities. The pathophysiology of CDG is attributed to disrupted protein glycosylation. However, the specific identities of the hypoglycosylated proteins responsible for most disease manifestations are unknown. In the nervous system, key proteins involved in neurotransmitter release, neuronal cell signaling, and cellular migration events are glycosylated, yet our understanding of the role of glycosylation in these processes is rudimentary. The genetic basis of CDG provides an opportunity to identify the neurobiological functions of glycosylation using model systems and glycoproteomics. Disruptions in glycosylation have also been implicated in the pathophysiology of complex neurologic and psychiatric diseases, including Alzheimer disease, amyotrophic lateral sclerosis (AML), and schizophrenia. The goal of my lab is to elucidate the biological roles of glycosylation in the brain and to develop novel therapeutic approaches to treating glycosylation-related disease manifestations. We currently focus on two major types of glycosylation, N-linked and O-linked.

N-linked glycosylation: Most CDG disrupt N-glycosylation. The single most common genetic cause of CDG results from biallelic mutations in PMM2. Patients with PMM2-CDG typically suffer from multi-systemic symptoms, including prominent and progressive neurological symptoms, such as intellectual disability, seizures, and cerebellar hypoplasia with resulting ataxia. Lifelong neurological impairment is the largest source of morbidity for PMM2-CDG patients. Currently, there is no effective treatment. We have several mouse models that recapitulate various aspects of the disease. Currently, we are employing mass spectrometry-based approaches to identify disrupted glycosylation for identification and validation of the molecular pathophysiology of disease mechanisms.

O-linked glycosylation: Our lab recently identified a new genetic cause of CDG, caused by biallelic mutations in GALNT2, which encodes a Golgi-localized glycosyltransferase that initiates mucin-type O-glycosylation. Patients with GALNT2-CDG exhibit global developmental delay, multi-focal treatment-resistant epilepsy, autistic features, and white matter changes on brain MRI. Currently, there is no effective treatment. We developed a mouse model for this disorder, with mice exhibiting spontaneous seizures and deficits across numerous behavioral and learning domains. Time-locked video EEG recordings identify seizures in the majority of these mice, which increase in prevalence as the mice age. Genetic dissection of the circuit suggests that molecular events in both excitatory and inhibitory neurons contribute to development of spontaneous seizures. These findings implicate a role of O-glycosylation in diverse neurological processes, including learning, memory, and neurotransmission. Glycoproteomic analysis of brains from these mice identified candidate glycoproteins and disrupted O-glycosites that likely underlie these abnormalities.

Links and key research papers

  • Polla DL, Edmondson AC, Duvet S, March ME, Sousa AB, Lehman A; CAUSES Study, Niyazov D, van Dijk F, Demirdas S, van Slegtenhorst MA, Kievit AJA, Schulz C, Armstrong L, Bi X, Rader DJ, Izumi K, Zackai EH, de Franco E, Jorge P, Huffels SC, Hommersom M, Ellard S, Lefeber DJ, Santani A, Hand NJ, van Bokhoven H, He M, de Brouwer APM: Bi-allelic variants in the ER quality-control mannosidase gene EDEM3 cause a congenital disorder of glycosylation. Am J Hum Genet 108: 1342-1349, Jul 2021.

  • Zilmer M, Edmondson AC, Khetarpal SA, Alesi V, Zaki MS, Rostasy K, Madsen CG, Lepri FR, Sinibaldi L, Cusmai R, Novelli A, Issa MY, Fenger CD, Abou Jamra R, Reutter H, Briuglia S, Agolini E, Hansen L, Petäjä-Repo UE, Hintze J, Raymond KM, Liedtke K, Stanley V, Musaev D, Gleeson JG, Vitali C, O’Brien WT, Gardella E, Rubboli G, Rader DJ, Schjoldager KT, Møller RS: Novel congenital disorder of O-linked glycosylation caused by GALNT2 loss of function. Brain 143: 1114-1126, Apr 2020.

  • Ligezka AN, Radenkovic S, Saraswat M, Garapati K, Ranatunga W, Krzysciak W, Yanaihara H, Preston G, Brucker W, McGovern RM, Reid JM, Cassiman D, Muthusamy K, Johnsen C, Mercimek-Andrews S, Larson A, Lam C, Edmondson AC, Ghesquière B, Witters P, Raymond K, Oglesbee D, Pandey A, Perlstein EO, Kozicz T, Morava E: Sorbitol Is a Severity Biomarker for PMM2-CDG with Therapeutic Implications. Ann Neurol 90(6): 887-900, Dec 2021.

Queenie Tan, M.D., Ph.D.

Medical Geneticist

Department of Clinical Genomics-Department of Laboratory Medicine and Pathology, Mayo Foundation for Medical Education and Research

Queenie Tan, M.D., Ph.D., is a board-certified clinical geneticist and clinical biochemical geneticist in the Department of Clinical Genomics at Mayo Clinic. Dr. Tan has expertise in the diagnosis and management of Inborn Errors of Metabolism, including Congenital Disorders of Glycosylation and Lysosomal Storage Diseases. She is also involved in the diagnostic workup, and the development of therapies for rare disorders. Dr. Tan did her PhD in Molecular Biology at Yale University, and completed training at Duke University Medical Center for General Pediatrics and Clinical Genetics, followed by Biochemical Genetics at University of Wisconsin at Madison. She is happy to be involved in the Minnesota Newborn Screening Advisory Committee.

Conditions treated

  • Genetic disorder
  • Metabolic disorders
  • Metabolic liver disease
  • Rare disorders

Interests

  • Newborn Screening
  • Inborn Errors of Metabolism
  • Lysosomal Storage Diseases
  • Glycogen Storage Diseases
  • Congenital Disorders of Glycosylation
  • Menkes Disease
  • Ultrarare Disorders

Links and key research papers