Prof. Xiaofeng JiaUSA
University of Maryland School of Medicine, Johns Hopkins University School of Medicine
Current Position
07/2019 to present The director of Neurosurgical Stem Cell Research and the director of the Translational Neuroengineering and Neuroscience Laboratory at the University of Maryland, Baltimore (UMB). He is a professor with tenure in the Department of Neurosurgery at the University of Maryland School of Medicine, Department of Orthopaedics, Anatomy Neurobiology, and a professor of the Department of Biomedical Engineering and Anesthesiology at Johns Hopkins University School of Medicine.
2016 to present The Section editor-in-chief of the International Journal of Molecular Sciences, Associate Editor for Frontiers in Immunology, Frontiers in Neuroscience, Frontiers in Neurology
Academic Experiences
2019 - 2022Secretary/Treasurer, Chair-elect, and Chair of Research Section Steering Committee, a member of the Discovery Steering Committee, and the Discovery Oversight Committee, the Society of Critical Care Medicine (SCCM)
Professional Experiences
2013 - 2024Chartered Member of the NIH study section for Acute Neural Injury & Epilepsy (ANIE) and an ad-hoc member of NIH study sections for CNNT; BMBI; ANIE; BINP; ZRG1 BDCN-K; ZRG1 CFS-N (80) S; and ETTN-10 Clinical Neurophysiology, Devices, Neuroprosthetics, and Biosensors Small Business Panel. He has been the invited ad hoc reviewer for the Department of Veterans Affairs, Research and Development Service (RR&D); National Natural Science Foundation of China (NSFC) Key Program and General Program of Department of Health Sciences panel meeting; EUROPEAN COMMISSION Research Executive Agency (REA); French Agence Nationale de la Recherche; United Kingdom Medical Research Council; Singapore National Medical Research Council; Hong Kong Research Grants Council; American Heart Association; Society of Critical Care Medicine; United Kingdom National Institute of Academic Anaesthesia (NIAA) et al.
Specialty & Expertise
Stem cell therapy; Peripheral nerve injury and regeneration; Brain recovery after cardiac arrest; Metabolic glycoengineering; Brain monitoring and therapeutic hypothermia; Spinal cord injury
About Me
He is an elected fellow of the American College of Critical Care Medicine (FCCM), and a member of the American Academy of Orthopaedic Surgeons (AAOS) (basic science), the American Association for Hand Surgery (AAHS). He is a recipient of the Annual Research Awards from AAHS in 2008 (among one of a total 3 awarded nationally), the 2009 Top 20 Scientific Exhibits and the 2008 Finalist of Best Overall Poster Award of AAOS, the CPCC Junior Investigator Travel Award from AHA, the Finalist of Research Citation Award from SCCM. His research work was highlighted as a significant advance based on animal research at the 65th American Academy of Neurology (AAN) Annual Meeting, 2013. He is a recipient of the Society of Critical Care Medicine 2021, 2022, and 2023 Star Research Achievement Award.
Presentation Information
Novel Stem Cell therapy to boost brain recovery after cardiac arrest
1108 16:20-16:30
Cerebrovascular/304B
Ischemic brain injury after cardiac arrest (CA) remains challenging with high mortality and disability and causes irreversible neurological dysfunction in over 90% of CA survivors. Neural stem cell (NSC) engrafting is an emerging therapeutic strategy with considerable promise that, however, is severely compromised by limited cell functionality after in vivo transplantation. Metabolic glycoengineering (MGE) employs sugar analogs to introduce functional groups to hNSC surface proteins to enhance cell adhesion, and, consequently, survival rates. To evaluate the impact of MGE on hNSC therapy for brain recovery after CA, we first developed the novel Ac5ManNTProp analog and optimized the treatment conditions and differentiation ability with hNSCs in vitro. To evaluate the TProp treatment on stem cell therapy and 4-week long-term outcomes after resuscitation, Wistar rats after 8 min-CA were randomly assigned to the control group, NSC group, and TProp-NSC group. To evaluate the effects of modulation Wnt signaling on functional outcomes after CA, adult Wistar rats were randomly treated with Wnt-inhibitor IWR-1 modified NSC and Wnt-promotor HLY78 modified NSC. Neurological deficit scores (NDS) were assessed daily for three days, and then weekly after resuscitation. Open-Field, Elevated Maze, and Tail Suspension tests were performed weekly to monitor behavioral progression. Cell migration was quantified using immunohistochemistry of brain tissues. MGE using the “TProp” monosaccharide analog stimulates the Wnt/β-catenin pathway, improves cell adhesion, and enhances neuronal differentiation in human NSCs in vitro thereby substantially increasing the therapeutic potential of these cells. For the first time, MGE significantly enhances NSC efficacy for treating ischemic brain injury after asphyxia CA in rats. In particular, neurological deficit scores and neurobehavioral tests, which showed improvement with "stand-alone" NSC therapy, achieved even greater efficacy when the therapeutic cells were pretreated with TProp. The TProp-NSC group exhibited significantly stronger neuroprotective functions including enhanced differentiation, synaptic plasticity, and reduced microglia recruitment; furthermore, Wnt pathway agonists and inhibitors demonstrated a pivotal role for Wnt signaling in these processes. MGE as a promising avenue for addressing current limitations associated with NSC transplantation via beneficially influencing neurite development and neural regeneration, offers enhanced therapeutic options to boost brain recovery following global ischemia. ACKNOWLEDGEMENT: This work was partially supported by R01NS125232 and R01NS110387 from the National Institute of Neurological Disorders and Stroke (both to Xiaofeng Jia).