Leadership Type: Scientific
A career history of fundamental discovery and translational research in immunology has guided Dr. Ware to identify new drug targets and develop novel therapeutics. Dr. Ware’s career in immunology and virology began in 1982 when he became a Professor at the University of California, Riverside’s Division of Biomedical Sciences. In 1996, he joined the La Jolla Institute for Immunology in San Diego as Head of the Division of Molecular Immunology. Professor Ware joined Sanford Burnham Prebys Medical Discovery Institute in 2010, serving as the Director of the Infectious and Inflammatory Disease Center and Adjunct Professor of Biology at the University of California at San Diego. He is currently the Director of the Laboratory of Molecular Immunology, which focuses on discovering and designing immunotherapeutics.
As an educator, he taught medical students immunology and virology. He trained over 60 postdoctoral fellows and graduate students who chose careers in research in academic and pharmaceutical science, patent law, or teaching.
Dr. Ware advises scientific panels and review boards for the National Institutes of Health and serves on the scientific advisor boards for the Allen Institute for Immunology and the Arthritis National Research Foundation. Scientific advisor with several biotechnology and pharmaceutical companies on immunotherapy for cancer and autoimmune diseases using innovative approaches to target discovery and drug development.
Dr. Ware’s research program is dedicated to unraveling the intricate intercellular communication pathways that govern immune responses. His work, which centers on cytokines in the Tumor Necrosis Factor (TNF) Superfamily, particularly those that regulate cell survival and death in response to viral pathogens, spans the domains of cancer,autoimmune and infectious diseases.
At Sanford Burnham Prebys, Dr. Ware is pivotal in promoting the translation of the faculty’s scientific discoveries. His efforts have led to the Institute’s reputation as a productive and preferred partner in collaborations with Pharma, including multi-year research and drug development projects with Eli Lilly and Avalo Therapeutics. His success translating fundamental knowledge into rational drug design has led to three novel therapeutics targeting inflammatory pathways, currently in clinical trials.
Education
1981-1982: T cell Immunology, Dana-Farber Cancer Institute of Harvard Medical School in Boston, MA. Tim Springer and Jack Strominger, advisors.
1979-1981: Biochemistry of Complement, University of Texas Health Science Center, San Antonio, TX. W Kolb, advisor
1974-1979: PhD in Molecular Biology and Biochemistry from the University of California, Irvine; Gale Granger, PhD mentor.
Honors and Recognition
Distinguished Fellow, American Association of Immunologists
Honorary Lifetime Membership Award International Cytokine and Interferon Society
Hans J. Muller-Eberhardt Memorial Lecture
Biotech All Star, San Diego Padres Awar
“Pillars of Immunology” discovery of the Lymphotoxin-b Receptor, published in Science
Outstanding Alumnus, Ayala School of Biological Sciences, University of California, Irvine
National MERIT Award R37 (10 years), National Institute of Allergy and Infectious Disease, NIH
National Research Service Award, NIH Postdoctoral Fellowship
Related Disease
Arthritis, Breast Cancer, Cancer, Crohn’s Disease (Colitis), Infectious Diseases, Inflammatory Bowel Disease, Inflammatory/Autoimmune Disease, Inherited Disorders, Leukemia/Lymphoma, Myeloma, Pathogen Invasion, Psoriasis, Systemic Lupus Erythematosus, Type 1 Diabetes
Research in the Laboratory of Molecular Immunology is directed at defining the intercellular communication pathways controlling immune responses. Our work is focused on the Tumor Necrosis Factor (TNF)-related cytokines in regulating decisions of cell survival and death, especially in responses to viral pathogens. Translational research is redirecting the communication networks of TNF superfamily to alter the course of autoimmune and infectious disease and cancer.
Carl Ware’s Research Report
Discovery of the TNF-LIGHT-LTαβ Network
The molecular elements of this cellular communication network were revealed with our discovery of Lymphotoxin-β and its formation of the trimeric heterocomplex with LTαβ1 and its signaling receptor, Lymphotoxin-β Receptor2. The LTβR revealed a new inter-cellular communication pathway that provides a key mechanism underlying the development and homeostasis of lymphoid organs. A second ligand we discovered, LIGHT (TNFSF14), is a novel ligand for the herpesvirus entry mediator (HVEM; TNFRSF14), and surprisingly, the LTβR3 heralding the concept that TNF, LTαβ, and LIGHT form an integrated signaling network thru distinct receptors controlling inflammation and host defense.4
LTβR Signaling in Host Defense and Immune Evasion
Our investigations into the mechanisms of virus evasion of the immune system revealed an essential role of the LTβR pathway in regulating the type 1 interferon response to cytomegalovirus5 and now recognized as a major defense against other pathogens. LTβR signals differentiate macrophages and stromal cells into IFN-producing cells. LTβR transcriptomics and proteomics datasets we generated revealed a novel constellation of anti-viral host defense mechanisms6. Importantly the role of the LTβR pathway to alter tissue microenvironments by differentiation of specialized stromal cells has implications for promoting effective immune responses to cancer.
Discovery of the HVEM-BTLA Immune Checkpoint

The LTBR-HVEM-BTLA Network in the TNF Superfamilies. Arrows indicate ligand-receptor binding (black), inhibitors (red arrow) and bidirectional signaling (dual arrowheads); HSV, herpes simplex virus; CMV, human cytomegalovirus. BTLA and CD160 are Ig Superfamily proteins. BTLA is an inhibitory checkpoint receptor; DcR3, decoy receptor 3 inhibits LIGHT binding to HVEM and LTBR.
The discovery that HVEM is a coreceptor for the immune checkpoint, B and T lymphocyte attenuator (BTLA), an Ig superfamily member, established a new paradigm in TNF Receptor signaling pathways 7. Additional investigations revealed the importance of the HVEM-BTLA system in limiting immune responses, including T cell help for B cell clonal expansion, antibody maturation, and secretion8. HVEM-BTLA also regulates control of the intestinal microbiome, limiting invasion of pathogenic bacteria and enhancing Treg cell homeostasis 9. The diverse roles of this pathway are seen in the loss of BTLA signaling from mutations in HVEM frequently present in B cell lymphomas10. Additional layers of immune regulators, CD160 and DcR3, control the LIGHT-HVEM-BTLA pathways, revealing this network as a key mechanism controlling immune homeostasis.
Appreciating the fundamental features of the TNF-LIGHT-LTab Network in effector and homeostasis mechanisms presents a target-rich resource for therapeutic intervention in autoimmunity, infection, and cancer11, 12.
Translational research and Immunotherapy
- 2021-current: Lead Scientific Advisor, Avalo Therapeutics
A neutralizing, fully human mAb (quisovalimab) to the proinflammatory cytokine LIGHT (TNFSF14) completed Phase I with an excellent safety profile and a Phase II trial establishing efficacy in COVID-19 pneumonia (NCT0441205)13. We identified elevated serum levels of LIGHT in hospitalized patients with COVID1914 spurring a randomized, double-blind, multicenter, proof-of-concept trial with adults hospitalized with COVID-19-associated pneumonia and mild to moderate ARDS15. The results established efficacy with a significant proportion of patients remaining alive and free of respiratory failure through day 28 after receiving quisovalimab, most pronounced in patients >60 years of age (76.5% vs. 47.1%, respectively; P = 0.042). These results and animal models validated LIGHT as a target for non-COVID inflammatory conditions, clinical trials ongoing in asthma (NCT05288504)12. - 2021-current: Principal Investigator, Avalo Therapeutics – Sanford Burnham Prebys collaboration
Bioengineered a first-in-class checkpoint agonist targeting BTLA immune checkpoint16 in preclinical development - 2019-current: Director and Principal Investigator, Fair Journey Biologics – Sanford Burnham Prebys collaboration
Immunotherapy for TNBC and PANC, in preclinical development - 2015-2022: Director and Lead Principal Investigator, LILLY-Sanford Burnham Prebys Collaboration in Autoimmunity
Collaborative research partnership with Eli Lilly involved target discovery and therapeutic development directed at immune regulators for autoimmune diseases. The collaboration produced three novel biologics currently in Phase I/2 trials (NCT03933943). The collaboration included a target discovery platform for T cell effector memory and NK cell immunomodulators. - 2015-2020: Lead Principal Investigator, Sanford Burnham Prebys – Capella Biosciences collaboration
Created a fully human mAb specific for membrane LIGHT (CBS001); phase I initiated (NCT05323110). - 2016-2020: Lead Scientific Investigator, Boehringer Ingelheim – Sanford Burnham Prebys Collaboration
Target discovery collaboration in inflammatory and fibrotic diseases6 - 2012-2014: Pfizer Innovation Center, Principal Investigator Bioengineering TNFR Superfamily in Autoimmune disease
- 1. Browning, J.L. et al. Lymphotoxin beta, a novel member of the TNF family that forms a heteromeric complex with lymphotoxin on the cell surface. Cell 72,847-856 (1993).
- 2. Crowe, P.D. et al. A lymphotoxin-beta-specific receptor. Science 264,707-710 (1994).
- 3. Mauri, D.N. et al. LIGHT, a new member of the TNF superfamily, and lymphotoxin alpha are ligands for herpesvirus entry mediator. Immunity 8,21-30 (1998).
- 4. Ward-Kavanagh, L.K., Lin, W.W., Sedy, J.R. & Ware, C.F. The TNF Receptor Superfamily in Co-stimulating and Co-inhibitory Responses. Immunity 44,1005-1019 (2016).
- 5. Schneider, K. et al. Lymphotoxin-mediated crosstalk between B cells and splenic stroma promotes the initial type I interferon response to cytomegalovirus. Cell Host Microbe 3,67-76 (2008).
- 6. Virgen-Slane, R. et al. Cutting Edge: The RNA-Binding Protein Ewing Sarcoma Is a Novel Modulator of Lymphotoxin beta Receptor Signaling. J Immunol 204,1085-1090 (2020).
- 7. Sedy, J.R. et al. B and T lymphocyte attenuator regulates T cell activation through interaction with herpesvirus entry mediator. Nat Immunol 6,90-98 (2005).
- 8. Mintz, M.A. et al. The HVEM-BTLA Axis Restrains T Cell Help to Germinal Center B Cells and Functions as a Cell-Extrinsic Suppressor in Lymphomagenesis. Immunity 51,310-323 e317 (2019).
- 9. Stienne, C. et al. Btla signaling in conventional and regulatory lymphocytes coordinately tempers humoral immunity in the intestinal mucosa. Cell reports 38,110553 (2022).
- 10. Sedy, J.R. & Ramezani-Rad, P. HVEM network signaling in cancer. Adv Cancer Res 142,145-186 (2019).
- 11. Croft, M., Benedict, C.A. & Ware, C.F. Clinical targeting of the TNF and TNFR superfamilies. Nat Rev Drug Discov 12,147-168 (2013).
- 12. Ware, C.F., Croft, M. & Neil, G.A. Realigning the LIGHT signaling network to control dysregulated inflammation. J Exp Med 219 (2022).
- 13. Zhang, M., Perrin, L. & Pardo, P. A Randomized Phase 1 Study to Assess the Safety and Pharmacokinetics of the Subcutaneously Injected Anti-LIGHT Antibody, SAR252067. Clin Pharmacol Drug Dev 6,292-301 (2017).
- 14. Perlin, D.S. et al. Levels of the TNF-Related Cytokine LIGHT Increase in Hospitalized COVID-19 Patients with Cytokine Release Syndrome and ARDS. mSphere 5 (2020).
- 15. Perlin, D.S. et al. Randomized, double-blind, controlled trial of human anti-LIGHT monoclonal antibody in COVID-19 acute respiratory distress syndrome. J Clin Invest 132 (2022).
- 16. Sedy, J.R. et al. A herpesvirus entry mediator mutein with selective agonist action for the inhibitory receptor B and T lymphocyte attenuator. J Biol Chem 292,21060-21070 (2017).
- Mar 25, 2025
Engineering antibodies with a novel fusion protein
Mar 25, 2025Fusing two immune system proteins leads to a new method of generating antibodies and may advance drug discovery.
- Nov 19, 2024
Protein superfamily crucial to the immune system experiences Broadway-style revival
Nov 19, 2024San Diego scientists note a resurgence of interest in research on protein family to find new drug candidates.
- Jun 1, 2023
Pumping the brakes on autoimmune disease
Jun 1, 2023New study describes the science behind an autoimmune disease treatment in a Phase 2 clinical trial Researchers at Sanford Burnham…
- Nov 2, 2022
Seeing the immune system in full color
Nov 2, 2022A new flow cytometer at the Institute will help researchers study the immune system with unprecedented resolution and speed. The…
- Apr 6, 2022
How our immune system controls gut microbes
Apr 6, 2022And how this relationship could help fight autoimmune diseases
- Jan 20, 2022
New COVID-19 drug passes phase 2 clinical trial
Jan 20, 2022The new treatment, developed by Avalo Therapeutics with Sanford Burnham Prebys researchers, can mitigate lung damage and improve survival in…
Select Publications
Showing 3 of 3Targeting the TNF and TNFR superfamilies in autoimmune disease and cancer.
Croft M, Salek-Ardakani S, Ware CF
Nat Rev Drug Discov 2024 Dec ;23(12):939-961Epitope topography of agonist antibodies to the checkpoint inhibitory receptor BTLA.
Cheung TC, Atwell S, Bafetti L, Cuenca PD, Froning K, Hendle J, Hickey M, Ho C, Huang J, Lieu R, Lim S, Lippner D, Obungu V, Ward-Kavanagh L, Weichert K, Ware CF, Vendel AC
Structure 2023 Aug 3 ;31(8):958-967.e3Randomized, double-blind, controlled trial of human anti-LIGHT monoclonal antibody in COVID-19 acute respiratory distress syndrome.
Perlin DS, Neil GA, Anderson C, Zafir-Lavie I, Raines S, Ware CF, Wilkins HJ
J Clin Invest 2022 Feb 1 ;132(3):Evan Y. Snyder earned his MD and PhD (in neuroscience) from the University of Pennsylvania in 1980 as a member of NIH’s Medical Scientist Training Program (MSTP). He had also studied psychology and linguistics at the University of Oxford. After moving to Boston in 1980, he completed residencies in pediatrics and neurology as well as a clinical fellowship in Neonatal-Perinatal Medicine at Children’s Hospital-Boston, Harvard Medical School. He also served as Chief Resident in Medicine (1984-1985) and Chief Resident in Neurology (1987) at Children’s Hospital-Boston. In 1989, he became an attending physician in the Department of Pediatrics (Division of Newborn Medicine) and Department of Neurology at Children’s Hospital-Boston, Harvard Medical School. From 1985-1991, concurrent with his clinical activities, he conducted postdoctoral research as a fellow in the Department of Genetics, Harvard Medical School. In 1992, Dr. Snyder was appointed an instructor in neurology (neonatology) at Harvard Medical School and was promoted to assistant professor in 1996. He maintained lab spaces in both Children’s Hospital-Boston and at Harvard Institutes of Medicine/Beth-Israel Deaconess Medical Center. In 2003, Dr. Snyder was recruited to Sanford Burnham Prebys as Professor and Director of the Program in Stem Cell and Regenerative Biology. He then inaugurated the Stem Cell Research Center (serving as its founding director) and initiated the Southern California Stem Cell Consortium. Dr. Snyder is a Fellow of the American Academy of Pediatrics (FAAP). He also received training in Philosophy and Linguistics at Oxford University.
Related Disease
Alzheimer’s Disease, Amyotrophic Lateral Sclerosis (Lou Gehrig’s Disease), Arthritis, Brain Cancer, Brain Injury, Breast Cancer, Cancer, Childhood Diseases, Congenital Disorders of Glycosylation, HIV-Associated Dementia, Huntington’s Disease, Multiple Sclerosis, Muscular Dystrophy, Neurodegenerative and Neuromuscular Diseases, Neurological and Psychiatric Disorders, Parkinson’s Disease, Peripheral Vascular Disease, Skin Cancer and Melanoma, Spinal Cord Injury, Stroke, Traumatic Injury
We believe the study of stem cell biology will provide insights into many areas: developmental biology, homeostasis in the normal adult, and recovery from injury. Indeed, past and current research has already produced data in these areas that would have been difficult or impossible via any other vehicle. We have engaged in a multidisciplinary approach, simultaneously exploring the basic biology of stem cells, their role throughout the lifetime of an individual, as well as their therapeutic potential. Taken together, these bodies of knowledge will glean the greatest benefit for scientists and, most importantly, for patients. All of our research to date has been preformed in animal models with the ultimate goal of bringing them to clinical trials as soon as possible. Stem cells offer an intriguing mix of controversy, discovery, and hope. Politicians are charged with dealing with the controversial facets of stem cells, as we prefer to focus our energy on their potential for discovery and hope.
The Snyder Lab studies stem cell biology, with the goal of understanding normal development, tissue homeostasis, and recovery from injury and disease. A major focus is neural stem cells (NSCs), which can self-renew and differentiate into neurons, astrocytes, and oligodendrocytes. These properties make NSCs ideal for repair of damage due to injury or disease, but they also make them susceptible to transformation into malignant cancers.
- Nov 11, 2024
Seven questions for FDA advisor Evan Snyder
Nov 11, 2024Sanford Burnham Prebys physician-scientist advises the FDA on cell-based therapeutics, tissue engineering and gene therapies.
- Jul 23, 2024
Mini lungs make major COVID-19 discoveries possible
Jul 23, 2024Scientists infect miniature lung organoids with the virus responsible for COVID-19, revealing new ways in which the infection spreads and…
- Jan 25, 2022
New CIRM grant to fund research internships for underrepresented high school students
Jan 25, 2022Thanks to a new grant awarded to Sanford Burnham Prebys by the California Institute for Regenerative Medicine (CIRM), 57 California…
- Nov 18, 2021
From child neurology to stem cells: An interview with Evan Snyder
Nov 18, 2021What do Evan Snyder and Sigmund Freud have in common? Both radically changed how we see the human brain. In…
- Jul 26, 2021
Biomarker could help diagnosis schizophrenia at an early age
Jul 26, 2021Research could lead to blood-based diagnostic test Scientists at Sanford Burnham Prebys have discovered how levels of a protein could…
- Feb 22, 2021
Evan Snyder named Fellow of the American Institute for Medical and Biological Engineering
Feb 22, 2021Renowned stem cell researcher recognized by his peers for seminal contributions to regenerative medicine The American Institute for Medical and…
Select Publications
Showing 3 of 3Probing the lithium-response pathway in hiPSCs implicates the phosphoregulatory set-point for a cytoskeletal modulator in bipolar pathogenesis.
Tobe BTD, Crain AM, Winquist AM, Calabrese B, Makihara H, Zhao WN, Lalonde J, Nakamura H, Konopaske G, Sidor M, Pernia CD, Yamashita N, Wada M, Inoue Y, Nakamura F, Sheridan SD, Logan RW, Brandel M, Wu D, Hunsberger J, Dorsett L, Duerr C, Basa RCB, McCarthy MJ, Udeshi ND, Mertins P, Carr SA, Rouleau GA, Mastrangelo L, Li J, Gutierrez GJ, Brill LM, Venizelos N, Chen G, Nye JS, Manji H, Price JH, McClung CA, Akiskal HS, Alda M, Chuang DM, Coyle JT, Liu Y, Teng YD, Ohshima T, Mikoshiba K, Sidman RL, Halpain S, Haggarty SJ, Goshima Y, Snyder EY
Proc Natl Acad Sci U S A 2017 May 30 ;114(22):E4462-E4471Proof of concept studies exploring the safety and functional activity of human parthenogenetic-derived neural stem cells for the treatment of Parkinson’s disease.
Gonzalez R, Garitaonandia I, Crain A, Poustovoitov M, Abramihina T, Noskov A, Jiang C, Morey R, Laurent LC, Elsworth JD, Snyder EY, Redmond DE Jr, Semechkin R
Cell Transplant 2015 ;24(4):681-90Neural stem cells implanted into MPTP-treated monkeys increase the size of endogenous tyrosine hydroxylase-positive cells found in the striatum: a return to control measures.
Bjugstad KB, Redmond DE Jr, Teng YD, Elsworth JD, Roth RH, Blanchard BC, Snyder EY, Sladek JR Jr
Cell Transplant 2005 ;14(4):183-92Pier Lorenzo Puri earned his MD at the University of Rome “la Sapienza” in 1991. Dr. Puri completed his internship in Internal Medicine at the hospital “Policlinico Umberto I” (Rome) from 1992 to 1997, and defended an experimental thesis on the vascular effects of angiotensin II to graduate as Specialist in Internal medicine at the University of Rome “la Sapienza” in 1997. During this time he was frequently working at the Freien University of Berlin, as visiting scientist at the Deprtment of Biochemistry and Molecular Biology, to perform experiments of protein and DNA microinjection in cultured cells. Dr. Puri trained as a post-doctoral fellow at the University of California San Diego (UCSD), in the department of Cell Biology, under the supervision of Dr. Wang, from 1997 to 2001. He was appointed as Staff Scientist at the Salk Institute (La Jolla) in 2001, and became an Assistant Telethon Scientist at the Dulbecco Telethon Institute in Rome in 2002. He was upgraded to Associate Telethon Scientist at the Dulbecco Telethon Institute in Rome since 2007 and became Senior Telethon Scientist, Dulbecco Telethon Institute, in 2012, but declined this position. Dr. Puri joined Sanford Burnham Prebys as an Assistant Professor in 2004. He has been promoted to Associate Professor in 2010 and full Professor in 2015. From 2008 to 2016 Dr. Puri served as Adjunct Professor of Pediatrics at the University of California, San Diego. From 2008 to 2013 Dr Puri was an Associate Member of Sanford Children’s Health Research Center. Dr Puri has been Director of the laboratory of Epigenetics and Regeneration at Fondazione S. Lucia, Roma, Italy, but stepped down this position since 2019.
Education
University of California San Diego, Postdoctoral, Department of Biology
University of Rome La Sapienza, PhD, Internal Medicine
University of Rome La Sapienza, MD, Internal Medicine
University of Rome La Sapienza, Undergraduate, Internal Medicine
Other Appointments
2020-2024: Member of the Science Advisory Board (SAB) European Commission-funded Consortium BIND (Brain Involvement In Dystrophinopathies)
2015-2019: Standing Member, NIH Study Section (SMEP)
2010-present: Member of Editorial Board of Skeletal Muscle
Related Disease
Aging-Related Diseases, Childhood Diseases, Molecular Biology, Muscular Dystrophy, Myopathy, Neurodegenerative and Neuromuscular Diseases, Rhabdomyosarcomas, Sarcopenia/Aging-Related Muscle Atrophy, Spinal Cord Injury, Transcription Factors
Phenomena or Processes
Adult/Multipotent Stem Cells, Aging, Cell Biology, Cell Cycle Progression, Cell Differentiation, Cell Signaling, Cellular Senescence, Development and Differentiation, Disease Therapies, DNA Damage Checkpoint Function, Epigenetics, Gene Regulation, Phosphorylation, Regenerative Biology, Signal Transduction, Transcriptional Regulation
Anatomical Systems and Sites
General Cell Biology, Musculoskeletal System
Research Models
Clinical and Transitional Research, Cultured Cell Lines, Human Adult/Somatic Stem Cells, Mouse Embryonic Stem Cells, Mouse Somatic Stem Cells, Primary Human Cells
Techniques and Technologies
Bioinformatics, Cellular and Molecular Imaging, Gene Expression, Genomics
Puri’s lab group investigates the molecular and epigenetic regulation of gene expression in skeletal muscle progenitors and other muscle-resident cell types (including fibro-adipogenic progenitors, cells from the inflammatory infiltrate, cellular components of neuro-muscular junctions) during physiological and pathological perturbations of skeletal muscle homeostasis.
- We use molecular, biochemical and epigenetic tools to understand structural and functional principles of the 3D genome organization that regulates gene expression during muscle regeneration and diseases.
- A topic of particular interest is the analysis of chromatin interactions that define the 3D genome organization and the identification of structural and functional interactions that regulate cell type-specific patterns of gene expression in response to cues released within the skeletal muscle regenerative environment in health and disease conditions, such as muscular dystrophies and other neuromuscular diseases.
- The knowledge derived from our studies is instrumental to elucidate the pathogenesis of muscular disorders and discover pharmacological interventions that promote muscle regeneration to repair diseased muscles.
- Current translational focus is devoted to:
- the study of the therapeutic potential of HDAC inhibitors for treatment of Duchenne Muscular Dystrophy (DMD)
- the identification of genome variants associated to DMD patient-specific patterns of expression of disease-modifier genes that can account for individual trends of disease progression beyond the common genetic deficiency of dystrophin
- the effect of dystrophin deficiency and restoration by gene therapy on 3D genome and transcriptional output of DMD myofibers; the therapeutic potential of extracellular vesicles released by fibro-adipogenic progenitors of DMD skeletal muscles exposed to HDACi.

Puri Lab
Pier Lorenzo Puri’s Research Report
Puri’s lab group investigates the molecular and epigenetic regulation of gene expression in skeletal muscle progenitors and other muscle-resident cell types (including fibro-adipogenic progenitors, cells from the inflammatory infiltrate, cellular components of neuro-muscular junctions) during physiological and pathological perturbations of skeletal muscle homeostasis.
We use molecular, biochemical and epigenetic tools to understand structural and functional principles of the 3D genome organization that regulates gene expression during muscle regeneration and diseases
A topic of particular interest is the analysis of chromatin interactions that define the 3D genome organization and the identification of structural and functional interactions that regulate cell type-specific patterns of gene expression in response to cues released within the skeletal muscle regenerative environment in health and disease conditions, such as muscular dystrophies and other neuromuscular diseases.
The knowledge derived from our studies is instrumental to elucidate the pathogenesis of muscular disorders and discover pharmacological interventions that promote muscle regeneration to repair diseased muscles
Current translational focus is devoted to:
- the study of the therapeutic potential of HDAC inhibitors for treatment of Duchenne Muscular Dystrophy (DMD)
- the identification of genome variants associated to DMD patient-specific patterns of expression of disease-modifier genes that can account for individual trends of disease progression beyond the common genetic deficiency of dystrophin
- the effect of dystrophin deficiency and restoration by gene therapy on 3D genome and transcriptional output of DMD myofibers; the therapeutic potential of extracellular vesicles released by fibro-adipogenic progenitors of DMD skeletal muscles exposed to HDACi.
1. Epigenetic regulation of skeletal myogenesis by histone acetyltransferases and deacetylases
Our earlier identification and characterization of acetyltransferases p300/CBP and PCAF, as transcriptional co-activators, and the histone deacetylases HDACs, as transcriptional co-repressors, of the myogenic determination factor MyoD1-3, respectively, inspired the experimental rationale toward exploiting pharmacological inhibition of HDAC to promote skeletal myogenesis.
- p300 is required for MyoD-dependent cell cycle arrest and muscle-specific gene transcription. Puri PL, Avantaggiati ML, Balsano C, Sang N, Graessmann A, Giordano A, Levrero M. EMBO J 1997 Jan 15 ;16(2):369-8
- Differential roles of p300 and PCAF acetyltransferases in muscle differentiation. Puri PL, Sartorelli V, Yang XJ, Hamamori Y, Ogryzko VV, Howard BH, Kedes L, Wang JY, Graessmann A, Nakatani Y, Levrero M. Mol Cell 1997 Dec ;1(1):35-45
- Class I histone deacetylases sequentially interact with MyoD and pRb during skeletal myogenesis. Puri PL, Iezzi S, Stiegler P, Chen TT, Schiltz RL, Muscat GE, Giordano A, Kedes L, Wang JY, Sartorelli V. Mol Cell 2001 Oct ;8(4):885-97
- Stage-specific modulation of skeletal myogenesis by inhibitors of nuclear deacetylases. Iezzi S, Cossu G, Nervi C, Sartorelli V, Puri PL. Proc Natl Acad Sci U S A 2002 May 28 ;99(11):7757-62
- Deacetylase inhibitors increase muscle cell size by promoting myoblast recruitment and fusion through induction of follistatin. Iezzi S, Di Padova M, Serra C, Caretti G, Simone C, Maklan E, Minetti G, Zhao P, Hoffman EP, Puri PL, Sartorelli V. Dev Cell 2004 May ;6(5):673-84
2. HDAC inhibitors as pharmacological intervention in DMD and other muscular dystrophies
Puri lab discovered that dystrophin-activated nNOS signalling controls HDAC2 activity, thereby revealing a previously unrecognized link between constitutive activation of HDAC2 and alteration of the epigenetic landscape of dystrophin-deficient muscles6,7. This discovery established the rationale for using HDAC inhibitors to counter the progression of Duchenne muscular dystrophy (DMD), by correcting aberrant HDAC activity in dystrophin-deficient muscles8-11.
- Functional and morphological recovery of dystrophic muscles in mice treated with deacetylase inhibitors. Minetti G. C., Colussi c., Adami R., Serra C., Mozzetta C., Parente V., Illi B., Fortuni S., Straino S., Gallinari P., Steinkhuler C., Capogrossi M., Sartorelli V., Bottinelli R., Gaetano C., Puri P.L. Nat. Med 2006 Oct ;12(10):1147-50.
- A Common Epigenetic Mechanism Underlies Nitric Oxide Donors and Histone Deacetylase Inhibitors Effect in Duchenne Muscular Dystrophy. Colussi C., Mozzetta C., Gurtner A. , Illi B., Straino S., Ragone G., Pescatori M., Zaccagnini G., Rosati G., Minetti G., Martelli F., Ricci E., Piaggio G., Gallinari P., Steinkulher C., Capogrossi M.C., Puri P.L*, Carlo Gaetano. Proc Natl Acad Sci U S A 2008 Dec 9 ;105(49):19183-7 *Corresponding author.
- Fibroadipogenic progenitors mediate the ability of HDAC inhibitors to promote regeneration in dystrophic muscles of young, but not old mdx mice. Mozzetta C., Consalvi S., Saccone V., Tierney M., Diamantini A., Mitchel K.J., Marazzi G., Borsellino G., Battistini L., Sassoon D., Sacco A., Puri P.L. EMBO Mol Med 2013 Apr ;5(4):626-39
- Histological effects of givinostat in boys with Duchenne muscular dystrophy. Neuromuscul Disord. Bettica P, Petrini S, D’Oria V, D’Amico A, Catteruccia M, Pane M, Sivo S, Magri F, Brajkovic S, Messina S, Vita GL, Gatti B, Moggio M, Puri PL, Rocchetti M, De Nicolao G, Vita G, Comi GP, Bertini E, Mercuri E. Neuromuscul Disord 2016 Oct ;26(10):643-649
- HDAC inhibitors tune miRNAs in extracellular vesicles of dystrophic muscle-resident mesenchymal cells. Sandonà M, Consalvi S, Tucciarone L, De Bardi M, Scimeca M, Angelini DF, Buffa V, D’Amico A, Bertini ES, Cazzaniga S, Bettica P, Bouché M, Bongiovanni A, Puri PL, Saccone V. EMBO Rep 2020 Sep 3 ;21(9):e50863
- Determinants of epigenetic resistance to HDAC inhibitors in dystrophic fibro-adipogenic progenitors. Consalvi S, Tucciarone L, Macrì E, De Bardi M, Picozza M, Salvatori I, Renzini A, Valente S, Mai A, Moresi V, Puri PL. EMBO Rep 2022 Jun 7 ;23(6):e54721
3. Control of chromatin structure in muscle cells by regeneration-induced signaling pathways
Upon the discovery and characterization of intracellular signaling pathways (i.e. p38, ERK and AKT cascades) that regulate muscle gene expression in myoblasts, in earlier studies during Puri’s postdoctoral training, Puri lab has revealed the mechanism by which muscle environmental cues are converted into epigenetic changes that regulate gene expression in healthy and diseased muscles, via extracellular signal-activated kinase targeting of chromatin-modifying enzymes. These studies provided the first evidence that regeneration activated p38 and AKT signaling cooperatively direct assembly and activation of histone acetyltransferases and chromatin remodeling SWI/SNF complex at myogenic loci in muscle progenitors12,13,15. Moreover, we discovered that regeneration-activated p38 targets Polycomb Repressory Complex (PCR2) at Pax7 locus to promote formation of repressive chromatin during satellite cells a ctivation14.
- p38 Pathway Targets SWI/SNF Chromatin Remodeling Complex to Muscle-Specific Loci. Simone C., Forcales S.V., Hill D., Imbalzano A.L., Latella L., and Puri P.L. Nat Genet 2004 Jul ;36(7):738-43
- Functional interdependence at the chromatin level between the MKK6/p38 and IGF1/Pi3K/AKT pathways during muscle differentiation. Serra C., Palacios D., Mozzetta C Forcales S., Ripani M., Morantte I., Jones D. Du K., Jahla U., Simone C., Puri P.L. Mol Cell 2007 Oct 26 ;28(2):200-13
- TNF/p38 alpha/Polycomb signalling to Pax7 locus in satellite cells links inflammation to the epigenetic control of muscle regeneration. Palacios D., Mozzetta C., Consalvi S., Caretti G., Saccone V., Proserpio V., Marquez V.E., Valente S., Mai A., Forcales S., Sartorelli V., Puri P.L. Cell Stem Cell 2010 Oct 8 ;7(4):455-69
- Signal dependent incorporation of MyoD-BAF60c into Brg1-based SWI/SNF chromatin-remodeling complex. Forcales S., Albini S., Giordani L., Malecova B., Cignolo L., Chernov A., Coutinho P., Saccone V., Consalvi S., Williams R., Wang K., Wu Z., Baranovskaya S., Miller A., Dilworth F., Puri P.L. EMBO J 2012 Jan 18 ;31(2):301-16
4. Epigenetic basis for activation of the myogenic program in ESCs and other pluripotent cell types
Puri lab studied the epigenetic determinants of human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) commitment to skeletal myogenesis, by investigating the hESC resistance to direct conversion into skeletal muscle upon ectopic expression of MyoD, which can otherwise reprogram somatic cells into the skeletal muscle lineage. These studies showed that hESC and hiPSC resistance to myogenic conversion is caused by the lack of expression of one structural component of the SWI/SNF chromatin remodelling complex – BAF60C – which is specifically induced in embryoid bodies13. Based on these studies, we have recently established a protocol of hESC-derived 3D contractile myospheres that offers the unprecedented opportunity to dissect and analyze the epigenetic dynamics that underlie the formation of skeletal muscles and to identify changes in the epigenome induced by contractile activity in healthy vs dystrophin-deficient myofibers16,20. We have also determined the identity of the general transcription factors implicated in the activation of skeletal myogenesis17, and we have discovered that replicative senescence is associated with acquisition of resistance to MYOD-mediated activation of muscle gene expression, caused by the constitutive activation of DNA damage repair (DDR) response that impairs cell cycle progression and MYOD activity18. Finally, our recent work has elucidated the mechanism by which MYOD regulates high-order chromatin interactions to define the tri-dimensional (3D) nuclear architecture for the activation of skeletal myogenesis during human somatic cell reprogramming into skeletal muscles19.
- Epigenetic reprogramming of human embryonic stem cells (hESCs) into skeletal muscle cells and generation of contractile myospheres. Albini S., Coutinho P., Malecova B., Giordani L., Savchenko A., Forcales S, Puri P.L. Cell Rep 2013 Mar 28;3(3):661-70
- TBP/TFIID-dependent activation of MyoD target genes in skeletal muscle cells. Malecova B., Dall’Agnese A., Madaro L., Gatto S., Coutinho Toto P., Albini S., Ryan T., Tora L., Puri PL. Elife 2016 Feb 25 ;5:e12534
- 18) DNA damage signaling mediates the functional antagonism between replicative senescence and terminal muscle differentiation. Latella L., Dall’Agnese A, Boscolo F., Nardoni C. Cosentino M., Lahm A, Sacco A., Puri P.L. Genes Dev 2017 Apr 1 ;31(7):648-659
- Transcription Factor-Directed Re-Wiring of Chromatin Architecture for Somatic Cell Nuclear Reprogramming Toward Trans-differentiation. Dall’Agnese A., Caputo L., Nicoletti C., di Iulio J., Schmitt A., Gatto S., Diao Y., Ye Z., Forcato M., Perera R., Bicciato S., Telenti A., Ren B., Puri P.L. Mol Cell 2019 Nov 7 ;76(3):453-472.e8
- Acute conversion of patient-derived Duchenne muscular dystrophy iPSC into myotubes reveals constitutive and inducible over-activation of TGFβ-dependent pro-fibrotic signaling. Caputo L, Granados A, Lenzi J, Rosa A, Ait-Si-Ali A, Puri PL, Albini S. Skelet Muscle 2020 May 2 ;10(1):13
5. Identification, functional, phenotypic and molecular characterization of muscle-interstitial cells – (the fibroadipogenic progenitors – FAPs) in healthy and diseased muscles.
Our work has elucidated the molecular determinants of the interplay between adult muscle stem cells and cellular components of their functional niche (i.e. FAPs), by identifying regulatory networks implicated in compensatory or pathogenic regeneration, and suggesting “disease stage-specific” responses to pharmacological treatment of neuromuscular disorders, such as DMD. Indeed, we have shown that HDACi promote compensatory regeneration and prevent fibro-adipogenic degeneration in mdx mice at early stages of diseases, by targeting a population of muscle interstitial cells – FAPs8 – and have identified a HDAC-regulated network that controls expression of myomiRs and alternative incorporation of BAF60 variants into SWI/SNF complexes to direct the pro-myogenic or fibro-adipogenic FAP activity21. Furthermore, we have recently identified specific subpopulations of FAPs (subFAPs) in physiological conditions and disease22 and we have discovered that specific subFAPs expand and adopt pathogenic phenotypes upon muscle denervation23 or in muscles of patients affected by type 2 diabetes24.
- HDAC-regulated myomiRs control BAF60 variant exchange and direct the functional phenotype of fibro-adipogenic progenitors in dystrophic muscles. Saccone V, Consalvi S, Giordani L, Mozzetta C, Barozzi I, Sandoná M, Ryan T, Rojas-Muñoz A, Madaro L, Fasanaro P, Borsellino G, De Bardi M, Frigè G, Termanini A, Sun X, Rossant J, Bruneau BG, Mercola M, Minucci S, Puri P.L. Genes Dev 2014 Apr 15 ;28(8):841-57
- Spectrum of cellular states within Fibro-Adipogenic Progenitors upon physiological and pathological perturbations of skeletal muscle. Malecova B., Gatto S., Etxaniz U, Passafaro M. Cortez A., Nicoletti C., Giordani L., Torcinaro A., De Bardi M., Bicciato S., De Santa F., Madaro L, Puri PL. Nat Commun 2018 Sep 10 ;9(1):3670
- Denervation-activated STAT3-IL6 signaling in fibro-adipogenic progenitors (FAPs) promotes myofibers atrophy and fibrosis. Madaro L, Passafaro M., Sala D., Etxaniz U., Lugarini F, Proietti D., Alfonsi MV, Nicoletti C., Gatto S., De Bardi M., Rojas-García R, Giordani L., Marinelli S., Pagliarini V., Sette C, Sacco A, Puri PL. Nat Cell Biol 2018 Aug ;20(8):917-927
- Human skeletal muscle CD90+ fibro-adipogenic progenitors are associated with muscle degeneration in type 2 diabetic patients. Farup J, Just J, de Paoli F, Lin L, Jensen JB, Billeskov T, Roman IS, Cömert C, Møller AB, Madaro L, Groppa E, Fred RG, Kampmann U, Gormsen LC, Pedersen SB, Bross P, Stevnsner T, Eldrup N, Pers TH, Rossi FMV, Puri PL, Jessen N. Cell Metab 2021 Nov 2 ;33(11):2201-2214.e11
- Mar 12, 2025
Meet the La Jolla researcher who helped discover new muscular dystrophy drug
Mar 12, 2025People in Your Neighborhood: Dr. Pier Lorenzo Puri of Sanford Burnham Prebys says he was ‘obsessed with doing something’ to…
- Nov 13, 2024
Decades of dedication led to FDA approval of a new treatment for Duchenne Muscular Dystrophy
Nov 13, 2024Nearly 30 years of discoveries by a Sanford Burnham Prebys scientist and collaborators lead to federal approval of the first…
- Dec 14, 2020
Our top 10 discoveries of 2020
Dec 14, 2020This year required dedication, patience and perseverance as we all adjusted to a new normal—and we’re proud that our scientists
- Sep 14, 2020
Scientists uncover a novel approach to treating Duchenne muscular dystrophy
Sep 14, 2020The study reveals a promising new therapeutic approach for the incurable muscle-wasting condition.
- Sep 10, 2019
How your DNA takes shape makes a big difference in your health
Sep 10, 2019The more we learn about our genome, the more mysteries arise. For example, how can people with the same disease-causing…
- Sep 8, 2016
MDA grant speeds research toward better treatments for Duchenne muscular dystrophy
Sep 8, 2016Pier Lorenzo Puri, MD, PhD, professor in the Development, Aging, and Regeneration Program, has focused his career on finding treatments to
Select Publications
Showing 3 of 3p38 pathway targets SWI-SNF chromatin-remodeling complex to muscle-specific loci.
Simone C, Forcales SV, Hill DA, Imbalzano AN, Latella L, Puri PL
Nat Genet 2004 Jul ;36(7):738-43Functional and morphological recovery of dystrophic muscles in mice treated with deacetylase inhibitors.
Minetti GC, Colussi C, Adami R, Serra C, Mozzetta C, Parente V, Fortuni S, Straino S, Sampaolesi M, Di Padova M, Illi B, Gallinari P, Steinkühler C, Capogrossi MC, Sartorelli V, Bottinelli R, Gaetano C, Puri PL
Nat Med 2006 Oct ;12(10):1147-50Transcription Factor-Directed Re-wiring of Chromatin Architecture for Somatic Cell Nuclear Reprogramming toward trans-Differentiation.
Dall’Agnese A, Caputo L, Nicoletti C, di Iulio J, Schmitt A, Gatto S, Diao Y, Ye Z, Forcato M, Perera R, Bicciato S, Telenti A, Ren B, Puri PL
Mol Cell 2019 Nov 7 ;76(3):453-472.e8Jamey Marth is a Professor at Sanford Burnham Prebys. He has also been Director of the Center for Nanomedicine at the University of California Santa Barbara and Professor in the Department of Molecular, Cellular, and Developmental Biology. Dr. Marth received a PhD degree in Pharmacology from the University of Washington where he trained in the laboratories of Roger M. Perlmutter and Edwin G. Krebs. Dr. Marth’s previous positions included Professor of Medical Genetics at the Biomedical Research Center, University of British Columbia; Professor of Cellular and Molecular Medicine at the University of California San Diego; and Investigator of the Howard Hughes Medical Institute.
Education
1987: PhD, University of Washington, Pharmacology
1984: BS, University of Oregon, Genetics and Chemistry
Honors and Recognition
2017: Karl Meyer Award, Society for Glycobiology
2009-2020: John Carbon Chair in Biochemistry and Molecular Biology
2009-2019: Duncan and Suzanne Mellichamp Chair in Systems Biology
2009: Julius Stone Lectureship Award: Society for Investigative Dermatology
1995-2009: Investigator Award, Howard Hughes Medical Institute
1991-1995: Faculty Scholarship, The Medical Research Council of Canada
Related Disease
Cancer, Colitis, Diabetes – General, Inflammatory/Autoimmune Disease, Sepsis
Dr. Marth is a molecular and cellular biologist specializing in diseases attributable to protein glycosylation. His education and training span molecular genetics, biochemistry, pharmacology, cell biology, immunology, hematology, developmental biology, microbiology, and glycobiology.
As an enzymatic process essential to cells, glycosylation produces saccharides linked by glycosidic bonds to proteins, lipids, and themselves, termed glycans. The vast majority of secreted and cell surface proteins are post-translationally modified by glycosylation during transit through the secretory pathway, termed glycoproteins. A widely used college level cell biology textbook authored by others includes glycans as one of the four main families of the organic molecules of all cells, with lipids, proteins, and nucleic acids and that together they compose the macromolecules and other assemblies of the cell. The structures of glycans (and lipids) are, however, synthesized by template-independent processes, rendering them hard to predict and study. Cells produce and regulate an abundant and diverse glycome of glycosidic linkages in which some of the biological information is decoded by one or more glycan-binding receptors, termed lectins.
Glycans and lectins represent a significant percentage of genes in the genomes of organisms, with several hundred present in mammals. Because glycan biosynthesis, diversification, and degradation rely upon corresponding gene and enzyme function, glycan function can be investigated similarly to other enzymatic and metabolic pathways, such protein phosphorylation. However, we and others found that intact organisms were typically required to discover the functions of protein glycosylation in mammals. His laboratory has focused on discovering the biological information contained within select glycosidic linkages of N- and O-glycans in determining the function and fate of discrete glycoproteins that further contribute to the pathogenesis of autoimmune disease, colitis, diabetes, and sepsis.
To understand the nature and extent of the information generated by glycosidic linkages, we have applied multiple molecular approaches to investigate protein glycosylation in mice and humans. In doing so, we have contributed to the development of enabling technologies with broad applicability, such as conditional mutagenesis by Cre-lox recombination in living animals to determine gene function with temporal and spatial selectivity. His laboratory also develops and studies experimental systems that may better represent real-world models of environmental factors that trigger acquired and common human diseases, results from which have been consistent with clinical findings of human patients. His laboratory includes interdisciplinary team-based collaborations that integrate expertise in immunology, infectious disease, hematology, and more recently, cancer, and is especially focused upon glycosidic linkages attached to the N- and O-glycans of glycoproteins.
The physiological systems regulated by protein glycosylation are broad even when comparing among sequential biosynthetic steps, and our findings continue to indicate the presence of undiscovered information of medical relevance residing in the glycan linkages of glycoproteins.
- Oct 31, 2024
Jamey Marth interviewed by The Scientist
Oct 31, 2024The Sanford Burnham Prebys scientist discussed the Cre-loxP recombination system, a mainstay genetic engineering technology.
- Sep 28, 2021
Jamey Marth awarded $13.5 million by NIH to investigate the pathogenesis and treatment of sepsis
Sep 28, 2021Sanford Burnham Prebys professor Jamey Marth, PhD, has been awarded $13.5 million from the National Heart, Lung, and Blood Institute to
- Jul 13, 2021
Study finds promising therapeutic target for colitis
Jul 13, 2021Neu3 controlled the emergence of disease in a model of human colitis An international research group, led by Jamey Marth, PhD,
- Oct 10, 2017
Jamey Marth honored for research linking glycans to diabetes, lupus, sepsis
Oct 10, 2017Jamey Marth, Ph.D., is the 2017 recipient of the Society for Glycobiology’s Karl Meyer Award. The international award is given…
- Jul 26, 2016
Team led by Jamey Marth awarded $12.8M to develop new ways to prevent sepsis
Jul 26, 2016A multidisciplinary team of scientists led by Jamey Marth, PhD, professor in the NCI-designated Cancer Center and director of UC Santa Barbara’s Center
- Oct 19, 2015
How proteins age
Oct 19, 2015SBP researchers and colleagues discover a mechanism that regulates the aging and abundance of secreted proteins. Physiological processes in the…
Select Publications
Showing 1 of 1Accelerated Aging and Clearance of Host Anti-inflammatory Enzymes by Discrete Pathogens Fuels Sepsis.
Yang WH, Heithoff DM, Aziz PV, Haslund-Gourley B, Westman JS, Narisawa S, Pinkerton AB, Millán JL, Nizet V, Mahan MJ, Marth JD
Cell Host Microbe 2018 Oct 10 ;24(4):500-513.e5Dr. Randal Kaufman previously served as professor of Biological Chemistry and Internal Medicine and Howard Hughes Medical Research Institute investigator at the University of Michigan Medical School. He received his PhD in pharmacology from Stanford University, where he studied gene amplification as a mechanism by which cells become resistant to anticancer agents. He was a Helen Hay Whitney fellow with Nobel Laureate Dr. Phillip Sharp at the Center for Cancer Research at the Massachusetts Institute of Technology (M.I.T.), where he developed gene transfer technologies based on gene amplification and expression in mammalian cells. He did his postdoctoral work at the Center for Cancer Research at M.I.T. In the 1980s, Dr. Kaufman’s experience with gene transfer and engineering led him to become a founding scientist at Genetics Institute Inc., where he engineered mammalian cells for high-level expression of therapeutic proteins, such as clotting factors that are now used to treat individuals with hemophilia. Dr. Kaufman joined Sanford Burnham Prebys in 2011.
Education
Postdoctoral, Center for Cancer Research, M.I.T.
PhD, Stanford University
BA, University of Colorado
Other Appointments
7/2011: Present Adjunct Professor, Department of Biological Chemistry, University of Michigan, Ann Arbor, MI
Honors and Recognition
2006: AAAS Fellow
2000: Distinguished Investigator Award-MI Hemophilia Society
1999: Investigator Recognition Award, International Society of Thrombosis and Haemostasis
1998: International Association Francaise Des Hemophiles Award
1993: Dr. Murray Thelin Award
Related Disease
Liver Diseases, Type 2 Diabetes
Phenomena or Processes
Protein Misfolding, The Unfolded Protein Response
The Kaufman lab is focused on understanding the fundamental mechanisms that regulate protein folding and the cellular responses to the accumulation of unfolded/misfolded proteins within the Endoplasmic Reticulum (ER). When proteins fail to fold correctly, they don’t work properly. More importantly, misfolded proteins accumulate with age and cause cellular toxicity, leading to almost every disease associated with aging. In many degenerative diseases, including neurological, metabolic, genetic, and inflammatory diseases, it’s thought that the accumulation of misfolded proteins leads to cellular dysfunction and death.
Dr. Kaufman’s research has focused for more than 30 years on mechanisms that regulate proper protein folding in the ER; this work contributed to the discovery of the UPR in the mid 1980s. The UPR pathways, mediated by PERK, IRE1, and ATF6, coordinate primarily an adaptive response. More recently, his research has focused on molecular mechanisms that establish the apoptotic program in response to protein misfolding in the ER, studies that have shed light on the mechanism by which cancer cells survive in a stressful environment.
Randal Kaufman’s Research Report
The major portion of our research is aimed at elucidating fundamental mechanisms that regulate protein folding and the cellular responses to the accumulation of unfolded protein within the (ER). Research into the fundamental processes that regulate protein synthesis and folding within the ER should have impact on the understanding of genetic diseases that result from protein folding defects.
Accumulation of unfolded/misfolded proteins within the ER induces an adaptive stress response known as the Unfolded Protein Response (UPR). The UPR signal is transduced from the ER lumen to cytoplasm and nucleus by three transmembrane proteins IRE1, ATF6, and PERK. UPR activation induces the expression of a family of basic leucine zipper-containing transcription factors that activate transcription of genes encoding functions to reduce the protein-folding load and increase the protein folding capacity of the ER. IRE1 is a serine/threonine protein kinase and endoribonuclease that signals transcriptional activation by initiating a novel splicing reaction on the mRNA encoding the transcription factor XBP1. UPR activation promotes trafficking of ATF6 from the ER to the Golgi where it is processed to yield a cytosolic fragment that is a potent transcriptional activator. In addition, the protein kinase PERK signals translational attenuation through phosphorylation of the alpha subunit of the eukaryotic translation initiation factor 2 (eIF2a) on serine residue 51. This phosphorylation attenuates translation of most cellular mRNAs but selectively induces translation of the transcription factor ATF4. We demonstrated that PERK/eIF2a signaling is essential for glucose-regulated insulin production by pancreatic beta cells, where defects in this pathway result in beta cell dysfunction and diabetes. The findings demonstrate an unprecedented link between glucose metabolism, mRNA translation, and protein folding and have implication in the treatment of diabetes. Future studies directed to elucidate the molecular logic for the UPR adaptive response will provide fundamental insight into numerous pathological conditions such as viral infection, cancer, inflammation, metabolic disease and atherosclerosis, and protein folding diseases such as Parkinson’s disease and Alzheimer’s disease.
- Dec 6, 2024
Three Sanford Burnham Prebys faculty members ranked among the world’s most influential scientists
Dec 6, 2024The publications of David A. Brenner, Randal J. Kaufman and Tariq M. Rana are among the most cited in the…
- Nov 15, 2023
Randal J. Kaufman among the world’s most influential scientists
Nov 15, 2023Over the last decade, the publications of Randal J. Kaufman are among the top 1% in the world by number…
- Dec 19, 2022
Misfolding proteins bring caution for gene therapies for haemophilia
Dec 19, 2022Researchers from Sanford Burnham Prebys, US, led by Dr Randal Kaufman, have found misfolded proteins in liver cells contribute to…
- Dec 5, 2022
Liver cancer study encourages caution with certain gene therapies
Dec 5, 2022A newly discovered link between protein misfolding and liver cancer could help improve gene therapy for hemophilia.
- Nov 21, 2022
Randal J. Kaufman among the world’s most highly cited researchers
Nov 21, 2022Over the last decade, the publications of Randal J. Kaufman are among the top 1% in the world by number…
- Mar 8, 2022
Randal Kaufman included in $12 million initiative to improve hemophilia treatment
Mar 8, 2022The new project will help researchers better understanding why current gene therapy treatments aren’t working.
Select Publications
Showing 3 of 3Chop/Ddit3 depletion in β cells alleviates ER stress and corrects hepatic steatosis in mice.
Yong J, Parekh VS, Reilly SM, Nayak J, Chen Z, Lebeaupin C, Jang I, Zhang J, Prakash TP, Sun H, Murray S, Guo S, Ayala JE, Satin LS, Saltiel AR, Kaufman RJ
Sci Transl Med 2021 Jul 28 ;13(604):Mechanisms, regulation and functions of the unfolded protein response.
Hetz C, Zhang K, Kaufman RJ
Nat Rev Mol Cell Biol 2020 Aug ;21(8):421-438Therapeutic opportunities for pancreatic β-cell ER stress in diabetes mellitus.
Yong J, Johnson JD, Arvan P, Han J, Kaufman RJ
Nat Rev Endocrinol 2021 Aug ;17(8):455-467Dr. Jackson brings many years of drug discovery and development experience to Prebys Center. Prior to joining Sanford Burnham Prebys in 2009, Dr. Jackson spent 15 years working within Johnson & Johnson’s pharmaceutical research organization. He managed cross-disciplinary teams of scientists focused on discovering chemical leads and clinical candidates directed at novel first-in-class drug targets. In 1999, he took on the role of vice president of discovery research at Johnson & Johnson’s La Jolla, California location, where he establish a state-of-the-art drugs discovery institute. As site head, Dr. Jackson oversaw all aspects of the new institute, from building design to hiring the staff of 300 scientists. In 2001 Dr. Jackson’s responsibility was expanded and as senior vice president of drug discovery at Johnson & Johnson Pharmaceutical Research and Development (U.S.) he was responsible > 800 hundred drug discovery scientists spread over five sites in the United States. Under his leadership, this organization advanced many clinical candidates into development across 5 multiple therapeutic areas including CNS, Pain, Immunology, CV disease. In 2005, Dr. Jackson was appointed President of ALZA Corporation, a large biotech company acquired by Johnson & Johnson that focused on drug delivery. As president of ALZA, he was responsible for all aspects of a 1,200 person research and development organization, successfully gaining regulatory approval for multiple drug delivery products that leverage the Oros technology and a first in class iontophoretic patch for the treatment of pain.
Prior to his industry career Dr. Jackson was an assistant professor at The Scripps Research Institute, where he published widely in the fields of cell biology and immunology. His work included identification of motifs that direct intracellular targeting of proteins to subcellular organelles, and the relevance of this and protein folding to antigen presentation and processing in the immune system. He received his PhD from the Department of Biochemistry at the University of Dundee in Scotland.
Career Highlights
- Managed (2009-2012) and then lead as P.I (2013-2014), Sanford Burnham’s NIH funded U54 grant Molecular libraries comprehensive screening center.
- Led the transformation of the Prebys center from a chemical biology and probe production organization to a full capable drug discovery operation generating first in class small molecule drug leads/NME’s.
- Instrumental in securing the Institutes first major multi-year, multi PI/project thematic translational collaboration with a pharmaceutical company.
- Instrumental in establishing the Florida Translational Research Program – a multi year contract from the department of health/State of Florida supporting Sanford Burnham Prebys translational drug discovery program in Orlando.
- Developed and executed “first of a kind” collaborative translational agreements with the Mayo clinic.
- Key member of the executive team that established a 10-year plan for the Institute that defined a path to sustainability. This plan was instrumental in securing an anonymous donation to the Institute of $275MM in January 2014.
- Established a pipeline of first in class therapeutics leads, the first product of this pipeline was licensed to Daiichi Sankyo in 2015.
- Key interface between the Institute and philanthropist Conrad Prebys, which resulted in a $100MM donation to the Institute in 2015.
- Instrumental is establishing a regional drug discovery effort for Alzheimer’s disease in collaboration with Alzheimer’s San Diego, Mayor’s office San Diego and SD County Supervisor.
Phenomena or Processes
Cell Biology
Techniques and Technologies
Drug Delivery, Drug Discovery
Dr. Jackson is senior vice president of drug discovery and development at Sanford Burnham Prebys, where he oversees the bicoastal operations of the Prebys Center, an approximately 80-person Drug Discovery enterprise embedded in the Institute. The center is equipped with state of the art ultra High Throughput Drug Screening (HTS) and lead discovery capabilities. His overall goal is to generate a pipeline of first in class small molecule therapeutics based on the break through discoveries on the molecular basis of disease made by investigators at the institute and collaborators.
Upon joining Sanford Burnham Prebys in 2009 Dr. Jackson managed and then lead as Principal Investigator, Sanford Burnham Prebys’ NIH-funded Molecular Libraries Probe comprehensive center which successfully executed over 100 HTS campaigns with collaborators across the Nation. Over a six year period the center generating >30 million screening data points and produced over 60 chemical probe reports published through the NCBI bookshelf. Since the Molecular libraries program ended the Prebys Center has continued to operate in a highly collaborative framework with investigators from many academic institutes and biopharmaceutical companies, it continues to conduct HTS screens against > 25 targets each year in search of drug leads to advance to therapeutics. Dr. Jackson is integrally involved in the center’s outreach and translational efforts that include over 35 ongoing collaborations, supported by grants, collaborations and contracts.
Dr. Jackson works closely with Sanford Burnham Prebys’ senior leadership to seek new avenues of support for preclinical and clinical drug development initiatives to enable the overall mission of the Institute to translate basic research to new products.
- Feb 12, 2025
Curebound awards two grants to Sanford Burnham Prebys scientists
Feb 12, 2025The San Diego-based philanthropic organization has awarded $43 million in cancer research to date.
- Sep 30, 2024
How AI can make drug discovery faster, better and cheaper
Sep 30, 2024In an essay, Michael R. Jackson, PhD, senior vice president for drug discovery and development at Sanford Burnham Prebys, explains.
- Sep 5, 2024
Acceleration by automation
Sep 5, 2024Increases in the scale and pace of research and drug discovery are being made possible by robotic automation.
- Aug 2, 2024
Sanford Burnham Prebys event explores the science behind addiction
Aug 2, 2024Scientists and clinicians from three local research institutions converged July 31 to discuss new ways to treat multiple addictions at…
- Jul 25, 2024
The Science Behind Addiction
Jul 25, 2024Scientists and clinicians from three local research institutions converge July 31 to discuss new ways to treat multiple addictions at…
- Nov 8, 2023
From tobacco to alcohol to opioids, Sanford Burnham Prebys researchers are pursuing novel leads and promising therapies to treat addiction
Nov 8, 2023Addiction is perhaps the most and least visible of public health crises in the US. Tens of millions of Americans…
Select Publications
Showing 3 of 3Cloning of cDNAs coding for rat hepatic microsomal UDP-glucuronyltransferases.
Jackson MR, McCarthy LR, Corser RB, Barr GC, Burchell B
Gene 1985 ;34(2-3):147-53The full length coding sequence of rat liver androsterone UDP-glucuronyltransferase cDNA and comparison with other members of this gene family.
Jackson MR, Burchell B
Nucleic Acids Res 1986 Jan 24 ;14(2):779-95Induction of UDP-glucuronyl transferase mRNA in embryonic chick livers by phenobarbital.
Jackson MR, Kennedy SM, Lown G, Burchell B
Biochem Pharmacol 1986 Apr 1 ;35(7):1191-8Dr. Freeze earned his PhD from the University of California, San Diego in 1976. Subsequently he held fellowships in Biology, Medicine and Neurosciences later joined the faculty at the same institution. In 1988 Dr. Freeze was recruited to Sanford Burnham Prebys.
Related Disease
Cancer, Congenital Disorders of Glycosylation, Crohn’s Disease (Colitis), Glycosylation-Related Disorders
Dr. Freeze’s research focuses on the pathology resulting from faulty glycosylation, the process of adding carbohydrate (sugar) chains to proteins and lipids. Carbohydrates are required for proper secretion and targeting of thousands of proteins – an often over looked fact of biology. He is driven by the search for novel therapeutics to treat patients with mutations leading to glycosylation defects called Congenital Disorders of Glycosylation (CDG).
Hudson Freeze’s Research Report
Glycosylation: An Essential Function
The entire cell surface is coated with sugars in complex chains that promote (or sometimes interfere) with cell-to-cell communication. These sugar chains are first attached to proteins deep inside the cell where they help them get into shape for their jobs. As the proteins percolate toward to cell surface and beyond, the sugar chains are sculpted for specific needs. This entire process, called glycosylation, recruits a force of more than 500 genes for this job. The Freeze lab works on several facets of glycosylation, all of them with an eye toward therapeutic applications for diseases that impair the functions of these critical genes.
Human Glycosylation Disorders
We focus is on a group of metabolic diseases called Congenital Disorders of Glycosylation (CDG). Today we know of defects in over 140 genes, well over double those known only 10 years ago. Patients with these diseases have highly variable mental and motor developmental delay, seizures, failure to grow, hypoglycemia (low blood sugar), clotting and digestion abnormalities and skeletal abnormalities to name just a few. These are rare disorders having a few thousand known patients worldwide, but it is likely that many remain undiagnosed, especially in developing countries.
Growing Awareness
Physicians are becoming more aware of glycosylation disorders in general, and basic scientists continue to discover sugar chains at the helm of many basic metabolic processes. Defective glycosylation is also known to cause 15 types of muscular dystrophy.

The explosive growth in the number of different diseases caused by defective glycosylation.
Working with CDG Kids
Rocket: We helped diagnose this young man over 10 years ago, and although he tragically passed away, his gentle face still reminds us of who we work for.

Rocket Williams reaches out to us. Learn more about the Rocket Fund.
Brianna: Sometimes our work leads to brighter outcomes as in the case of Brianna who we met over 20 years ago and treated with a simple therapy.

Flourishing, Brianna is now training to become a veterinarian.
Our Collaborations
The Freeze lab not only identifies new glycosylation disorders, but also tries to understand how these defects cause the disease manifestations. Defects occur in genes that activate and transport sugars, assemble them into glycans and remodel them. Some also traffic and distribute the glycosylation machinery within cells. Ongoing collaborations with academic physicians provide a steady flow of new patients for analysis. Since very few laboratories in the United States work on CDG, we are developing new molecular diagnostic methods to handle the increasing number of patients. Increased awareness of CDG in the medical community generated expanded government funding that allies us with 10 medical centers in the US seeking additional patients to study their natural history, develop biomarkers and test emerging therapies. Those avenues along with the help of generous philanthropic support, enables us to extend efforts to supplement the depleted glycosylation pathways in patients.

In this image and film clip above, Harrison Ford poses a few questions for us.
- Apr 1, 2025
How basic science fuels economic gain
Apr 1, 2025A recent story in The New York Times describes how new and changed federal policies pose long-term risk to science and the…
- Jan 2, 2025
Hudson Freeze finds life in hot water
Jan 2, 2025Hudson Freeze, PhD, has a starring cameo in Veritasium’s video profile of Kary Mullis, PhD, an American biochemist who shared the 1993 Nobel Prize in…
- Nov 11, 2024
Honoring Hud Freeze and colleagues for literally writing the book on glycobiology
Nov 11, 2024Glycobiology is the study of the structure, biosynthesis and biology of glycans — carbohydrates or sugars that cover the surfaces…
- Mar 28, 2024
At a symposium on rare diseases, smiles were in abundance
Mar 28, 2024Since 2010, we have organized an annual scientific and family conference at Sanford Burnham Prebys that covers multiple rare diseases.
- Mar 7, 2024
Sharing science and stories at Rare Disease Day Symposium
Mar 7, 2024The Sanford Burnham Prebys Rare Disease Day Symposium brought patients, families, physicians, scientists, industry experts and advocates together with a…
- Jan 3, 2024
Hudson Freeze appointed to Distinguished Endowed Chair, thanks to gift from grateful supporter
Jan 3, 2024An endowed chair is among the highest forms of recognition for a faculty member’s research, teaching and service.
- Our inspiration for CDG research Watch | Scientists in the Hudson Freeze lab talk about their inspiration for doing Congenital Glycosylation Disorder (CDG) research
- Para-equestrian with rare disorder meets Olympian in San Diego Watch | For 20-year-old Morgan Hill, it’s a dream come true – the chance to meet an Olympic medalist and lifelong hero
- “Miracle:” Uridine at 2 days and 5.5 months Watch | Uridine at 2 days and 5.5 months
Select Publications
Showing 3 of 3N-Glycanase 1 Transcriptionally Regulates Aquaporins Independent of Its Enzymatic Activity.
Tambe MA, Ng BG, Freeze HH
Cell Rep 2019 Dec 24 ;29(13):4620-4631.e4SLC35A2-CDG: Functional characterization, expanded molecular, clinical, and biochemical phenotypes of 30 unreported Individuals.
Ng BG, Sosicka P, Agadi S, Almannai M, Bacino CA, Barone R, Botto LD, Burton JE, Carlston C, Chung BH, Cohen JS, Coman D, Dipple KM, Dorrani N, Dobyns WB, Elias AF, Epstein L, Gahl WA, Garozzo D, Hammer TB, Haven J, Héron D, Herzog M, Hoganson GE, Hunter JM, Jain M, Juusola J, Lakhani S, Lee H, Lee J, Lewis K, Longo N, Lourenço CM, Mak CCY, McKnight D, Mendelsohn BA, Mignot C, Mirzaa G, Mitchell W, Muhle H, Nelson SF, Olczak M, Palmer CGS, Partikian A, Patterson MC, Pierson TM, Quinonez SC, Regan BM, Ross ME, Guillen Sacoto MJ, Scaglia F, Scheffer IE, Segal D, Singhal NS, Striano P, Sturiale L, Symonds JD, Tang S, Vilain E, Willis M, Wolfe LA, Yang H, Yano S, Powis Z, Suchy SF, Rosenfeld JA, Edmondson AC, Grunewald S, Freeze HH
Hum Mutat 2019 Jul ;40(7):908-925Pathogenic Variants in Fucokinase Cause a Congenital Disorder of Glycosylation.
Ng BG, Rosenfeld JA, Emrick L, Jain M, Burrage LC, Lee B, Undiagnosed Diseases Network, Craigen WJ, Bearden DR, Graham BH, Freeze HH
Am J Hum Genet 2018 Dec 6 ;103(6):1030-1037Dr. Commisso obtained his PhD at the University of Toronto in the Department of Molecular Genetics and completed his postdoctoral training at New York University School of Medicine. Dr. Commisso made the seminal discovery that Ras-mutant cancer cells use macropinocytosis as an amino acid supply pathway. His laboratory’s research interests are centered on elucidating the underlying mechanisms of how metabolic stress influences the tumor ecosystem in pancreatic cancer. The lab is particularly interested in identifying metabolic adaptations and dependencies that contribute to tumor progression and can be targeted to develop novel therapeutic modalities for this disease.
Related Disease
Cancer, Colorectal Cancer, Lung Cancer, Pancreatic Cancer
Research in the Commisso lab is focused on biological discoveries that have the potential to lead to novel therapeutic strategies for cancer. Of particular interest to our laboratory are Ras-driven cancers, such as pancreatic cancer, which are extremely aggressive and are in urgent need of new and innovative therapies. The biological process that we study in the lab is called macropinocytosis, a fluid-phase form of bulk endocytic uptake, which we have linked to cancer cell metabolism in Ras-mutated tumors.
Cosimo Commisso’s Research Report
Macropinocytosis is an endocytic mechanism of fluid-phase uptake that produces large intracellular vesicles known as macropinosomes. Macropinosomes are heterogeneous in size and shape and serve to internalize large volumes of extracellular fluid along with the associated membrane. In transformed cells, macropinocytosis is stimulated by oncogenes, such as Ras. Ras proteins are small, membrane-localized GTPases that are activated in response to growth factors and they regulate a variety of outputs, including cell proliferation, survival and invasion. Gain-of-function mutations in Ras-encoding genes cause Ras proteins to be trapped in their active state, leading to the constitutive activation of downstream pathways. The functional consequences of macropinocytosis stimulation in mutant Ras-expressing cells were unknown prior to our work. We have linked macropinocytic uptake in Ras-transformed cells to nutrient delivery and amino acid supply (Commisso et al., 2013). We demonstrated that the inhibition of this nutrient delivery pathway selectively compromises growth of Ras-driven tumors. With the long-term goal of specifically targeting such tumors, we have recently developed stream-lined methodology to detect and grade macropinocytosis in tumor tissue (Commisso et al., 2014). Our work was important for two main reasons. First, cancer cells are dependent on amino acids, such as glutamine, for their growth and survival. Therefore, the targeting of these amino acid supply pathways, such as macropinocytosis, represents a promising strategy in developing anti-cancer therapeutics. Second, macropinocytosis is emerging as a mechanism of entry for a variety of therapeutic agents, such as nanoparticles. Hence, identifying that this uptake pathway is active in Ras-driven tumors may have an impact on how these tumors are treated. The complete understanding of the functional significance of Ras-induced macropinocytosis to carcinogenesis and treatment ultimately depends on having a firm grasp of how this process is regulated and on the ability to specifically control it. To this end, a major research focus of the lab is to advance our understanding of the molecular pathways that drive macropinocytosis, which could lead to the identification of new molecular targets whose inhibition would restrain tumor growth and enhancers that could be manipulated to dial-up the uptake process in drug delivery strategies. Additional research interests in the lab include nutrient sensing pathways that are active in the context of macropinocytic uptake and macropinosome maturation, the process that leads to active protein catabolism within the tumor cell.
- Dec 5, 2024
Controlling cancer cells’ gluttony for glutamine
Dec 5, 2024Scientists discover new insights about how pancreatic cancer cells adapt when nutrients and other resources are scarce.
- Oct 18, 2024
Two Sanford Burnham Prebys scientists selected for American Cancer Society postdoctoral fellowships
Oct 18, 2024The funds will support Alicia Llorente Lope and Ambroise Manceau who study breast and pancreatic cancer.
- Jul 17, 2024
Sanford Burnham Prebys announces new faculty recruit and two faculty promotions
Jul 17, 2024Douglas Sheffler was named as a new associate professor at Sanford Burnham Prebys, while Cosimo Commisso and Nicholas Cosford garnered…
- May 22, 2024
Pancreatic cancer symposium celebrates 10th anniversary in San Diego
May 22, 2024The 2024 PancWest Symposium brought more than 120 scientists to the Sanford Burnham Prebys campus in San Diego to discuss…
- Oct 6, 2023
Copycat nutrient leaves pancreatic tumors starving
Oct 6, 2023Treatment approach tricks tumors with a compound that mimics a critical nutrient.
- Sep 5, 2023
US Department of Defense backs Cosimo Commisso’s pancreatic cancer research
Sep 5, 2023Cosimo Commisso, PhD, has received a grant from the Department of Defense for $1 million to advance the research of…
Select Publications
Showing 3 of 3Glutamine mimicry suppresses tumor progression through asparagine metabolism in pancreatic ductal adenocarcinoma.
Recouvreux MV, Grenier SF, Zhang Y, Esparza E, Lambies G, Galapate CM, Maganti S, Duong-Polk K, Bhullar D, Naeem R, Scott DA, Lowy AM, Tiriac H, Commisso C
Nat Cancer 2024 Jan ;5(1):100-113Macropinocytosis in Cancer-Associated Fibroblasts Is Dependent on CaMKK2/ARHGEF2 Signaling and Functions to Support Tumor and Stromal Cell Fitness.
Zhang Y, Recouvreux MV, Jung M, Galenkamp KMO, Li Y, Zagnitko O, Scott DA, Lowy AM, Commisso C
Cancer Discov 2021 Jul ;11(7):1808-1825Golgi Acidification by NHE7 Regulates Cytosolic pH Homeostasis in Pancreatic Cancer Cells.
Galenkamp KMO, Sosicka P, Jung M, Recouvreux MV, Zhang Y, Moldenhauer MR, Brandi G, Freeze HH, Commisso C
Cancer Discov 2020 Jun ;10(6):822-835Dr. Bradley received her doctorate from the University of California Berkeley, in 1981 in studies of CD4 T cell subsets that regulate humoral immune responses. Her work on the regulation of CD4 T cells continued during her postdoctoral training at The Oregon Primate Research Center and at the University of California, San Diego where she was appointed Assistant Research Professor in 1991. It was at this time she developed NIH sponsored her research program on CD4 T cells and discovered the key associations between migration and function. She joined The Scripps Research institute as an Assistant Professor in 1996 where she expanded her work on CD4 T cells into the arena of autoimmunity and discovered the essential role of the cytokine, interleukin-7, in the regulation of CD4 cell homeostasis.
She joined the Sidney Kimmel Cancer Center in 2001 as an Associate Professor, and was promoted to Professor in 2005. She joined Sanford Burnham Prebys as a Professor in the Infectious and Inflammatory Diseases Center in 2009. Dr. Bradley is recognized as a key contributor in the field of CD4 T cell biology, is an invited speaker at many national and international meetings, and serves on several study sections for the NIH as well as the Welcome Trust, Medical Research Council, and the JDRF.
Related Disease
Cancer, Infectious Diseases, Skin Cancer and Melanoma
Phenomena or Processes
Adaptive Immunity, Cell Signaling, Infectious Disease Processes, Inflammation
Anatomical Systems and Sites
Immune System and Inflammation
The research program in the Bradley lab is focused on understanding the regulation of T lymphocytes in virus infections where the immune response results in viral clearance and the development of immunologic memory, and in chronic virus infections where the ongoing immune response leads to viral persistence and immune dysregulation. They are guided by these studies to interrogate cellular mechanisms that can be modulated to promote better responses not only to virus infections, but also to relieve immune inhibition in the setting of cancer where T cells progressively lose function. Understanding adhesion mechanisms underlie the ability of T cells to become localized in tissues to eradicate infections and tumors is a key underpinning of their work.
The Bradley lab’s current focus is on two molecules that can function on T cells to initiate the processes that lead to their migration from the blood into tissue, CD44 and PSGL-1 (P-selectin glycoprotein-1). They have found that both of these receptors have key regulatory functions that are independent of their roles in migration. These proteins regulate the magnitude of T cell responses, as well as the survival and memory formation by T cells by different mechanisms, affecting processes in the T cell and stromal cell compartments.
Their ongoing studies of these immune checkpoint regulators using in vivo models indicate that they are promising therapeutic targets to enhance T cell responses to infections and cancer as well as to inhibit T cell responses in autoimmunity. They are therefore pursuing translational studies with the UCSD Moores Cancer center to analyze their regulation in human T cells in an effort to enhance patient responses to their tumors using in vivo modeling. In addition working to develop biologics for treatment of patients with autoimmunity and cancer.
- Aug 19, 2024
Women in Science event at Sanford Burnham Prebys examines how female faculty members navigate research careers
Aug 19, 2024Topics at the event included work/life balance, caregiving and family obligations, and gender disparities in academic rank at research and…
- Mar 26, 2024
Melanoma’s mysteries revealed at Sanford Burnham Prebys
Mar 26, 2024Cancer Center open house welcomes San Diego community to learn the latest about melanoma research.
- May 3, 2023
Reviving exhausted T cells to tackle immunotherapy-resistant cancers
May 3, 2023A new approach to immunotherapy could help overcome treatment resistance in cancer. When the cells of our immune system are…
- Mar 20, 2023
Sanford Burnham Prebys researchers awarded Curebound grants
Mar 20, 2023Each year, Sanford Burnham Prebys joins Padres Pedal the Cause, an annual fundraising event that raises money for Curebound which…
- Mar 19, 2018
Cancer immunology symposium highlights hot area in cancer research
Mar 19, 2018The Cancer Immunology and Tumor Microenvironment Symposium held at Sanford Burnham Prebys Medical Discovery Institute (SPB) on March 8, 2018…
- May 16, 2017
What SBP Scientists are Researching to Battle Skin Cancer
May 16, 2017Skin cancer is one of the most common of all cancers, and melanoma accounts for about 1 percent of skin…
Select Publications
Showing 1 of 1PSGL-1 Is an Immune Checkpoint Regulator that Promotes T Cell Exhaustion.
Tinoco R, Carrette F, Barraza ML, Otero DC, Magaña J, Bosenberg MW, Swain SL, Bradley LM
Immunity 2016 May 17 ;44(5):1190-203