Faculty Mentors
The faculty listed on this page have expressed special interest in the Propel program. As you look for potential postdoc mentors, consider how faculty research interests align with your own.
We recommend starting with the faculty listed on this page and then expanding your search to other faculty across the university. For School of Medicine faculty, browse SoM Departments or find details about individual faculty members in the School of Medicine via Community Academic Profiles (CAP).
Faculty members sign up here.
Name | Primary Appointment | Research |
---|---|---|
Alyce Adams https://profiles.stanford.edu/alyce-adams | Health Policy | health services and policy research focused on addressing healthcare disparities in chronic disease outcomes |
Nima Aghaeepour https://nalab.stanford.edu/team/nima-aghaeepour/ | Anesthesiology | We are a machine learning and artificial intelligence laboratory, broadly interested in all aspects of medicine. |
Ash Alizadeh https://med.stanford.edu/profiles/arash-alizadeh | UTL. Medicine, Oncology | Cancer Genomics, Data Science, Liquid Biopsies, Immuno-Oncology |
Nicolas Altemose https://profiles.stanford.edu/altemose | Genetics | The Altemose Lab develops new experimental and analytical tools to study how chromatin proteins organize and regulate complex regions of the human genome. |
Cristina Alvira https://profiles.stanford.edu/cristina-alvira | Associate Professor of Pediatrcis | Identification of molecular mechanisms that direct postnatal lung development, repair and regeneration using single cell transcriptomics, primary cell culture, and genetic mouse models and translating this knowledge to human disease. |
Jason Andrews https://med.stanford.edu/profiles/jason-andrews#publications | Medicine | Our laboratory aims to develop and test innovative approaches to the diagnosis, treatment and control of infectious diseases in resource-limited settings. We draw upon multiple fields including mathematical modeling, microbial genetics, field epidemiology, and statistical inference to work on challenging problems in infectious diseases, with an emphasis on tuberculosis and tropical diseases. |
Rebecca Aslakson https://profiles.stanford.edu/rebecca-aslakson | Medicine - MCL; I am joint appointed 50% in Department of Medicine and 50% in Department of Anesthesiology, Perioperative, and Pain Medicine | I am a clinical and health services researcher and all my work involves improving delivery of effective and equitable palliative care, particularly to ICU and perioperative populations. My research work is usually mixed methods using patient-centered research approaches involving significant stakeholder engagement. Database design and management, clinical trial/clinical research management, qualitative research methods, and community-based participatory research skills are fundamental to my research work. |
Themistocles (Tim) Assimes https://profiles.stanford.edu/tim_assimes | Department of Medicine, School of Medicine | I conduct molecular epidemiology studies of cardiovascular diseases with special emphasis on coronary artery disease and cardiometabolic risk factors. We use multiple -omics technologies to conduct our research in both small specialized datasets as well as large biobanks. |
Jeff Axelrod https://profiles.stanford.edu/jeffrey-axelrod | Pathology | We study developmental morphogenetic patterning events using genetic, molecular, cell biological and mathematical approaches. We use the fruitfly Drosophila melanogaster and the mouse as model systems in these investigations. Our primary focus is on Planar Cell Polarity (PCP), a signaling mechanism that orients cells within tissues and organs. Our studies aim to understand the mechanisms responsible for providing, interpreting and executing orientation signals. These signals are important not only in development, but also are of central importance to a variety of developmental defects and disease states, including neural tube closure defects, congenital deafness, ciliary dyskinesia and cancer, in which PCP mechanisms are mis-regulated. |
Jade Benjamin-Chung https://profiles.stanford.edu/jadebc | Epidemiology & Population Health | Our lab's mission is to improve population health by creating high quality evidence about what health interventions work in whom and where, when, and how to implement them. Most of our research is focused on infectious diseases, including malaria, diarrhea, soil-transmitted helminths, and influenza. Our focus is on improving the health of vulnerable populations from low-resource settings, both domestically and internationally. We use a variety of epidemiologic, computational, and statistical methods, including causal inference and machine learning methods, in pursuit of our mission. |
Daniel Bernstein https://profiles.stanford.edu/daniel-bernstein | Pediatrics (Cardiology) | Our lab addresses the mechanisms by which mutations in contractile proteins cause diseases such as hypertrophic and dilated cardiomyopathy. We utilize induced pluripotent stem cell cardiomyocytes which are CRISPR-edited with mutations, as well as human myocardial samples and mouse models. Our particular focus is on metabolic/energetic alterations, assessed by integrating metabolomics and transciptomics, as well as alterations in mitochondrial structure and function. |
Vivek Bhalla https://profiles.stanford.edu/vivek-bhalla | Medicine | In the Bhalla laboratory we use both in vitro and in vivo approaches for several projects related to the role of the kidney in health, diabetes, and hypertension. (1) Diabetic kidney disease is the most common form of chronic kidney disease in the world, yet no curative therapy is available. Studies of the susceptibility of diabetic kidney disease led to the discovery of differential regulation of endothelial-specific molecule-1, Esm-1 (endocan) in susceptible strains of mice. Esm-1 is a secreted proteoglycan that is enriched in glomerular endothelium and decreases albuminuria in the setting of diabetes and other inflammatory diseases. Ongoing rescue and deletion experiments explore the role of Esm-1 in diabetes and diabetic kidney disease. Studies using single cell analysis inform the biology of select Esm-1(+) vs. Esm-1(-) endothelial cells, and we are currently developing genetic tools to better interrogate the role of local vs. systemic Esm-1 as this would inform potential therapeutic strategies. We also a locus within the gene as a molecular switch for regulation of transcription. (2) Investigation of the mechanisms of hypertension in the setting of obesity and insulin resistance using renal tubular epithelial insulin receptor deletion challenged the role of insulin in the hypertension of obesity, insulin resistance, and the metabolic syndrome. These studies also shed light on the role of insulin in control of glucose reabsorption via SGLT2. Ongoing studies focus on molecular mechanisms of insulin-regulated SGLT2 and its contrast with insulin resistant pathways in other cell types and tissues. Our current work is focused on post-translational modifications of SGLT2 and their contribution to SGLT2 expression and function. (3) A major regulator of sodium reabsorption, Clcnkb, is mutated in humans with Barrter's syndrome, Type 3. However, surprisingly little is known about the regulation of this channel in health and disease. Ongoing experiments include the study of post-translational regulation of Clcnkb surface expression and activity, including glycosylation. (4) Inhibition of sodium reabsorption using diuretics is a mainstay of therapy for hypertension and edema-forming states. Study on the consequences of diuretic therapy using tubular morphometry and single cell approaches, combined balance studies have led to additional work on mechanisms of tubular remodeling in vivo. |
Nidhi Bhutani https://profiles.stanford.edu/nidhi-bhutani | Orthopedic Surgery | We are a multidisciplinary team with expertise in epigenetics, mouse models, clinical OA, stem cells and biomaterials for tissue engineering applications. Our research interests broadly encompass the molecular mechanisms regulating development, regeneration and repair of the musculoskeleton with a focus on epigenetics. This has led to our recent discoveries regarding epigenetic regulators playing a key role in the initiation and progression of Osteoarthritis (published in Science Translational Medicine, April 2020; Trends in Pharmacol. Sci, 2020). Additionally, I am interested in applying embryonic and induced pluripotent stem cells towards musculoskeletal tissue engineering along with using cutting-edge single cell techniques to understand distinct and rare subpopulations that orchestrate regeneration and disease. |
Helen M. Blau https://profiles.stanford.edu/intranet/helen-blau | MicroImmuno – Baxter Lab | Our research encompasses cell and molecular approaches to regenerative medicine for acquired and inherited diseases. A central interest is the elucidation of the mechanisms that underlie changes in muscle stem cell function in aging, muscular dystrophy, and diabetes. This knowledge is key to our understanding of stem cell self-renewal and expansion during tissue regeneration with a view toward medical applications for increasing muscle function and extending healthspan. A hallmark of our work is the development of interdisciplinary technologies that enable novel fundamental insights and drug discovery. We employ stem cells, murine models, bioengineering techniques, and bioinformatics approaches. Our current major focus is on the regeneration of muscle. In December 2020 we published a study in Science showing that decreasing the abundance of the prostaglandin PGE2 can trigger premature muscle aging in young mice and increasing its abundance rejuvenates the muscles of old mice. We further showed that PGE2 levels can be readily modulated by administration of a small molecule that inhibits 15-PGDH, the enzyme that degrades PGE2 leading to a marked increase in muscle mass and strength. We have multiple ongoing research projects expanding upon these initial findings. |
Catherine Blish https://profiles.stanford.edu/catherine-blish | Medicine/Infectious Diseases | Human immunology, infectious diseases, innate immunology, systems immunology. We seek to apply single cell technologies like single cell RNA-seq, single cell ATAC-seq, and proteomic profiling with CyTOF to understand host responses to infectious diseases, including SARS-CoV-2, HIV, dengue virus, influenza virus, and tuberculosis. |
Alistair Boettiger https://profiles.stanford.edu/alistair-boettiger?tab=bio | Assistant Professor, Developmental Biology | Understanding gene regulation, genome structure and epigenetic memory using super-resolution microscopy, high-throughput imaging, computational analysis, modeling. |
Melissa Bondy https://profiles.stanford.edu/melissa-bondy | Epidemiology and Population Health | My research interests are broad from molecular/genetic epidemiology to health disparities and population studies. I mostly focus on cancer but interested in many different areas. I have access to large genomic data sets and other data including EHR and data sets in the Center for Population Health. |
Onn Brandman https://profiles.stanford.edu/onn-brandman | Biochemistry | protein quality control, biophysics, stress responses, |
Paul Buckmaster https://profiles.stanford.edu/paul-buckmaster | Comparative Medicine | Temporal lobe epilepsy is common, frequently refractory to treatment, and devastating to those affected. Our long-term goal is to better understand the pathophysiological mechanisms of this disease so that rational and effective therapies can be developed. Key questions are how do brains develop epilepsy, and in epileptic brains how do seizures start? We use in vitro and in vivo electrophysiological techniques and anatomical methods to address these key questions. |
Marion Buckwalter https://profiles.stanford.edu/marion-buckwalter | Neurology and Neurological Sciences | My group works on neuroinflamation after stroke and how it affects short- and long-term outcomes. We have studies in both mice and humans. |
Andres Cardenas https://profiles.stanford.edu/Andres-Cardenas | Epidemiology and Population Health | Our group investigates prenatal and early-life determinants of health and disease. We conduct epidemiological analyses of human cohorts to investigate chemical (e.g. metals, endocrine disruptors) and non-chemical stressors (e.g. adversity and discrimination) and their relationships to human health and development. We use computational and bioinformatics approaches to study epigenetic and DNA methylation biomarkers in humans. Our group also has a special interest in human aging and epigenetic biomarkers of aging. |
Suzan Carmichael https://profiles.stanford.edu/suzan-carmichael | DEPARTMENT OF PEDIATRICS, SCHOOL OF MEDICINE | Our team focuses on population-based studies of maternal and infant health. In particular, we are currently focused on understanding contributors to severe maternal morbidity and mortality and disparities in these outcomes, including social/structural determinants, quality of care, maternal weight, clinical pathways, and how all of these aspects intersect. We are also interested in risk factors and care related to stillbirth and structural congenital anomalies. |
Charles Chan https://profiles.stanford.edu/charles-chan | Surgery | Stem Cell Niches, Aging and Rejuvenation, Skeletal Stem Cells, Cartilage Regeneration |
Howard Y Chang https://profiles.stanford.edu/howard-chang | Dermatology | Our research is focused on mechanisms that coordinate the activities of large number of genes in cell fate control. We made a series of discoveries that introduced the important and pervasive roles of long noncoding RNAs in biological regulation. My group has substantial experience in epigenetics and RNA biology, including invention of new methods for epigenomic profiling, map RNA occupancy on chromatin, and define RNA structures genome-wide. My group pioneered methods to identify key regulators of large-scale transcriptional programs; these methods have been highly fruitful for studies of development, cancer, and aging. The long term goal of my laboratory is to decipher the regulatory information in the human genome for disease diagnosis and therapy. |
Akshay Chaudhari https://profiles.stanford.edu/akshay-chaudhari | Radiology | The mission of our group is to develop and validate new techniques to improve the acquisition, interpretation, and ultimately, value of medical imaging technologies. Research in this group combines new artificial intelligence techniques with classical engineering and physics knowledge, with a particular emphasis on solving problems where only limited labeled datasets are available. We explore techniques revolving around semi/self/un-supervised learning, representation learning, multi-modality learning, and model explainability for image analysis and inverse problems. Projects in the group have a focus on improving the efficacy and value of healthcare, and subsequently, entail collaborations amongst faculty and students with technical and clinical expertise. A major principle of the MIMI group is to enable open-sourcing of algorithms and datasets to the broader research community. |
James Chen https://med.stanford.edu/profiles/james-chen | Chemical and Systems Biology | The Chen Lab explores the crossroads of chemistry, developmental biology, and cancer biology. They have discovered small-molecule and genetic regulators of Hedgehog signaling through high-throughput screens, and they have used light-activatable antisense oligonucleotides and zebrafish models to study the genetic programs that regulate mesoderm development. Current research interests include optogenetic regulators of developmental signaling pathways, small-molecule modulators of cancer stem cell metabolism, spermatogenesis, and male contraception. |
Alan Cheng https://profiles.stanford.edu/alan-cheng | Otolaryngology-HNS | Our research focuses on studying mechanisms of inner ear hair cell development and regeneration. We use a combination of in vitro and in vivo models studying mouse and human inner ear, with the goal of regenerating functional hair cells to restore inner ear function. Techniques used include molecular biology, confocal and live cell imagine, tissue culture, animal surgery, and auditory and vestibular physiology. |
Gheorghe Chistol https://profiles.stanford.edu/gheorghe-chistol | CHemical and Systems Biology | Chistol Lab is using real-time single-molecule imaging to: (i) study how eukaryotes replicate/repair their DNA, (ii) dissect molecular mechanisms involved in maintaining large/complex genomes, and (iii) understand how massive multi-subunit molecular machines like the replisome are regulated. |
Katrin Chua https://profiles.stanford.edu/katrin-chua | Medicine | Our research explores how molecular mechanisms of chromatin regulation and nuclear signaling impact on diverse epigenetic programs and how these mechanisms are de-regulated in aging, cancer, and other disease processes. We focus on the mammalian SIRT6 and SIRT7 genes, members of the evolutionarily conserved “Sirtuin” gene family that has key roles in regulating lifespan, metabolism, and stress resistance in many organisms. We also have a longstanding interest in understanding cellular senescence and its roles in aging and age-related pathologies. We combine biochemical, cellular, mouse model, and multi-omic methodologies. |
Karlene Cimprich https://profiles.stanford.edu/karlene-cimprich | Chemical and Systems Biology | The Cimprich lab is interested in many aspects of genome instability, particularly as they relate to DNA replication. They seek to understand the impact of replication stress signaling and DNA damage tolerance pathways on genome instability as well as the genome instability resulting from interactions between replication and transcription. Their work has also established the central importance of DNA-RNA hybrids or R-loops as a major endogenous source of replication stress and genome instability, and they are interested in understanding the mechanisms by which cells respond to and process R-loops. A combination of approaches, including molecular and cell biology, biochemistry and genomics, are used to study these topics, primarily in human cells. |
Tom Clandinin https://profiles.stanford.edu/thomas-clandinin | Neurobiology | systems and molecular neuroscience, neurodegeneration |
Shoa Clarke https://profiles.stanford.edu/shoa | Medicine | We integrate genetics, epidemiology, and data science to understand cardiovascular disease risk and motivate early preventive strategies. |
Le Cong https://profiles.stanford.edu/186687 | Pathology, Genetics | Our group are developing technology for genome editing and single-cell genomics, using computational approaches inspired by data science. His group has a focus on using these tools to study immunological and infectious diseases. |
David Cornfield https://profiles.stanford.edu/david-cornfield | Pediatrics | 1. Lung development; 2. Regulation of pulmonary vascular tone; 3. oxygen sensing; 4. single cell RNA-seq; 5. Preterm Labor |
Jerry Crabtree Gerald R Crabtree | Pathology | Our laboratory is interested in developing a new class of molecules to treat cancer that cause the cancer driver to directly activate precise and powerful pathways of programmed cell death. These molecules use a dominant gain-of-function mechanism that allows them to kill cancer cells even in the presence of multiple mutated cancer drivers and other oncogenic pathways. These molecules which we call TCIP for Transcriptional/Epigenetic Chemical Inducers of Proxmity, use the fundamental mechanism of chemically induced proximity to rewire cellular circuitry(Gourisankar, Krokhotin et al Nature 2023). For an overview of this work see "Flipping a Switch and Making Cancers Self-Destruct" by Gina Kolata in the New York Times. |
Jeremy Dahl https://profiles.stanford.edu/intranet/jeremy-dahl?releaseVersion=10.5.1 | Radiology | Diagnostic ultrasound and ultrasound engineering including the following technical areas: beamforming, image reconstruction, ultrasound imaging systems, image quality improvement, aberration & aberration correction, sound speed estimation, Doppler imaging, signal processing and ultrasound molecular imaging. Application areas include a broad array of clinical uses, but generally include: liver & abdominal imaging, NAFLD, cardiovascular imaging, maternal-fetal-medicine, and cancer imaging. |
Heike Daldrup-Link https://profiles.stanford.edu/heike-daldrup-link | Radiology | Cancer Imaging, Pediatric Imaging, Stem Cell Imaging, Molecular Imaging |
Rhiju Das https://profiles.stanford.edu/rhiju-das | Biochemistry | RNA biochemistry, RNA structural biology, RNA synthetic biology |
Vinicio De Jesus Perez https://profiles.stanford.edu/vinicio-de-jesus-perez | Associate Professor of Medicine | pulmonary hypertension, right heart failure, genetics, wnt signaling, vascular biology, drug toxicity |
Luis de Lecea https://profiles.stanford.edu/luis-de-lecea | Professor. Psychiatry and Behavioral Sciences | Our lab is interested in fundamental questions in the neuroscience of brain states: Why do we sleep? How does the brain regulate sleep and wakefulness? How does the brain decide it is appropriate to sleep or wake up? We also study how dysfunction of the arousal systems lead to anxiety, depression and addiction. |
Tushar Desai https://med.stanford.edu/profiles/tushar-desai#bio | Pulmonary, Allergy and Critical Care Medicine | My lab studies lung development, lung stem cells, and diseases like lung cancer and pulmonary fibrosis. We also develop new technologies for staining tissues to be able to investigate and compare healthy and diseased lungs to reveal the underlying basis for the disease and devise new treatments. The experimental assays we primarily use include multiplexed antibody and in situ hybridization, single cell RNA sequencing, mouse genetic models, spheroid, organoid and lung slice cultures, and confocal microscopy. |
Joseph DeSimone https://profiles.stanford.edu/joseph-desimone | Radiology and Chemical Engineering | Our lab develops new digital polymer 3D printing capabilities and materials for applications in human health. We have a major focus on translational medicine in areas including: new medical devices opportunities; microneedle-based vaccine platform development; precision delivery of therapies (molecular and cellular) and vaccines; device-assisted, targeted drug delivery; molecular monitoring; and digital treatment planning using novel pediatric therapeutic devices. |
Renumathy Dhanasekaran https://profiles.stanford.edu/renumathy-dhanasekaran | Assistant Professor | The overall goal of my research is to understand the molecular pathogenesis of liver cancer and identify biologically relevant prognostic biomarkers and molecular targets for therapy in patients with hepatocellular carcinoma (HCC). My current ongoing projects focus on these areas- 1. Drivers of tumor dormancy and recurrence of hepatocellular carcinoma (HCC) 2. Drivers of metastatic tumor progression in HCC 3. Prognostic gene signature for HCC 4. Glycoproteomic biomarkers of HCC 5. Using tumor cell free DNA (cfDNA) as biomarkers for HCC 6. Tumor progression of HCC in non alcoholic steatohepatitis (NASH) |
Max Diehn https://profiles.stanford.edu/maximilian-diehn | Cancer Institute & Radiation Oncology | Liquid Biopsies, Lung Cancer, Early Detection, Immuno-Oncology |
Dylan Dodd https://med.stanford.edu/profiles/dylan-dodd | Assistant Professor of Pathology | One of the key ways that the gut microbiome impacts human health is through the production of bioactive metabolites. By understanding how microbes produce these molecules, we aim to develop new approaches to promote human health and treat disease. Our laboratory employs bacterial genetics, metabolomics, and gnotobiotic mouse colonization to uncover the chemistry that underlies host-microbe interactions in the gut. |
Shaul Druckmann https://profiles.stanford.edu/shaul-druckmann | Neurobiology | Computational neuroscience |
Alfredo Dubra https://profiles.stanford.edu/alfredo-dubra | Ophthalmology | he Dubra lab is part of the Byers Eye Institute and the Ophthalmology Department at Stanford University. We seek to reveal ocular, vascular, neurodegenerative and systemic diseases through novel optical ophthalmic imaging technologies. Our work is motivated by the personal interactions with research study volunteers and patients that we have been fortunate to have worked with. Our current projects are focused on instrumentation for non-invasive visualization of retinal structure and function at a microscopic scale. We pursue this through a multidisciplinary approach that integrates optics, computer science, vision science, electrical engineering and other engineering disciplines. The major focus of our work is technology translation and dissemination which is furthered through collaboration with a remarkable group of scientists and clinicians at various institutions. This collaborative approach allows us to collectively develop methods for early diagnosis, precise monitoring of disease progression, evaluation of novel treatments, and improved understanding of disease mechanisms. |
Laramie Duncan https://profiles.stanford.edu/laramie-duncan | Psychiatry and Behavioral Sciences | Dr. Duncan’s work is at the intersection of psychology, statistical genetics, and neuroscience. The Duncan Lab use massive datasets and primarily computational approaches to identify risk factors and mechanisms contributing to mental health problems like schizophrenia and post-traumatic stress disorder (PTSD). Translational work in the lab leverages biomarkers and human postmortem brain tissue to advance the ultimate goal of building more rational approaches to classification, prevention, and treatment of psychiatric disorders. |
Gozde Durmus NTRL | Radiology, Molecular Imaging Division (MIPS) | diagnostics, cell sorting, biosensors, tissue engineering, regenerative medicine |
Elizabeth Egan https://profiles.stanford.edu/elizabeth-egan | Pediatrics | The research in our laboratory focuses on Plasmodium falciparum malaria, which is one of the leading causes of childhood disease globally. We are a basic science laboratory interested in host-pathogen interactions in malaria, with the goal of advancing our understanding of this complex disease and discovering new approaches to treatment and prevention. We are particularly focused on elucidating the role of human red blood cells in malaria infection, as P. falciparum is an obligate intracellular parasite of human red cells. Many of our studies rely on various genetic approaches in primary hematopoietic stem cells to generate mutants and differentiate them to mature red cells by ex-vivo erythropoiesis. Ongoing research projects range from discovering novel host factors for P. falciparum, understanding the mechanisms underlying red cell factors in malaria parasite invasion, the impact of infection on human bone marrow development, and the influence of natural red cell variation on malaria parasite fitness and pathogenicity. |
Edgar Engleman https://profiles.stanford.edu/edgar-engleman | Professor of Pathology | Our lab is comprised of postdocs and grad students from different disciplines – currently immunology, cancer biology, and bioengineering. We seek to decipher the role of the immune system in disease and therapy - ie, identify the immune cells and molecules responsible for disease development - and then target them with novel immunotherapeutic approaches with the ultimate goal of curing the disease. The majority of our projects are designed to discover new immunotherapies for cancer, but we also seek to discover new therapies for autoimmune disease and organ transplant rejection. We utilize all available methods (in vivo, in vitro, in silico) to identify candidate cells and genes/molecules of interest. We rely on mouse models of disease to test their roles in vivo, and study human tissues to validate our findings. Our discoveries provided the basis for the first FDA approved immunotherapy – the Sipuleucel-T (Provenge) vaccine that was approved in 2010 for the treatment of advanced prostate cancer. In 2015, my lab discovered a promising strategy that reprograms immunosuppressive tumor associated myeloid cells into immunostimulatory antigen presenting cells, in situ, that entered clinical trials in 2021 for the treatment of several different cancers. |
Jesse Engreitz https://profiles.stanford.edu/jesse-engreitz | Genetics | The Engreitz Lab is mapping the regulatory wiring of the genome to understand the genetic basis of heart diseases. This wiring includes millions of enhancers that tune gene expression in the thousands of cell types in the body. Yet, it has been unclear which enhancers regulate which genes — a massive and complex network that rewires in each cell type. To understand this network, we invent new genomics tools combining CRISPR and single-cell approaches; dissect molecular mechanisms of enhancer-gene communication; build computational models to map genome regulation; and apply these tools to connect human genetic variants to biological mechanisms of disease. |
Daniel Ennis https://profiles.stanford.edu/daniel-ennis | Department of Radiology | Dr. Ennis directs the Cardiac Magnetic Resonance (CMR) Group that develops translational cardiac and cardiovascular MRI techniques to improve clinical care. Current research projects focus on: 1) characterizing several biomarkers to detect the cardiomyopathy associated with Duchenne Muscular Dystrophy; 2) developing MRI methods and a computational modeling framework to estimate changes in passive ventricular stiffness in patients with Heart Failure with Preserved Ejection Fraction (HFpEF); and 3) developing MRI methods and a computational modeling framework to estimate changes in atrial fibrosis for patients with atrial fibrillation (AFib). Our group is also very interested in further developing MRI methods that measure Flow, Motion, Diffusion and Perfusion with particular emphasis on clever approaches to optimal gradient waveform design. Overall goals are to improve measurement accuracy and precision and to reduce the image acquisition times for cardiac MRI exams. We also work on MRI safety, especially with regards to patients with implanted pacemakers and ICDs. |
Neir Eshel https://profiles.stanford.edu/neir-eshel | Psychiatry & Behavioral Sciences | The STAAR Lab is a dynamic new neuroscience lab in Stanford’s Psychiatry Department, led by Neir Eshel, MD, PhD and embedded in the world-class scientific environment of Dr. Rob Malenka’s group. We are looking to hire curious and ambitious postdocs beginning summer 2022. Lab projects focus on the neural circuitry of addiction and aggressive behaviors, using optogenetics, in vivo imaging, electrophysiology, and sophisticated machine learning/artificial intelligence analyses of animal behavior. There are ample opportunities for career development and clinical exposure based on candidate interest. Compensation and benefits are highly competitive. The ideal postdoctoral candidate has an MD and/or PhD in neuroscience or related field and extensive experience with rodent neuroscience. Excellent analytical skills, e.g., Python & Matlab, are strongly preferred. An expert data analyst may be considered even without animal experience. We are strongly committed to diversity and inclusion. |
Titilola Falasinnu https://profiles.stanford.edu/titilola-falasinnu | Division of Immunology and Rheumatology | Characterizing the epidemiology of acute and chronic pain in rheumatology and in the general population using electronic health records, survey data and registries. |
Ryann Fame https://profiles.stanford.edu/ryann-fame | Neurosurgery- UTL | Early neural progenitors respond to extrinsic cues that maintain and support their potency. These stem/ progenitor cells are in direct contact with the cerebrospinal fluid (CSF), which acts as part of their niche. Our research program encompasses the early neural stem cell niche, neural tube closure, CSF, metabolism, and cortical neuronal development. We are dedicated to broad collaboration focused on translating an understanding of neurodevelopment and CSF biology into regenerative strategies. |
Heidi M Feldman https://profiles.stanford.edu/heidi-feldman | School of Medicine, Department of Pediatrics | My research focuses on the development of language in children, particularly children born preterm. My current study, called PRELUDES, interrogates the interactions of features of the social environment and characteristics of white matter pathways in the brain in the development of language. Participants are born at < 32 weeks gestation, receive a clinical MRI, diffusion MRI and quantitative T1 image before they are discharged home. We assess the social environment using questionnaires and specialized recording equipment that children wear for a day at home. The main outcome measures are collected when the children reach 18 months of age. This study is just beginning to enroll. Post-doctoral fellows would be involved in obtaining MRI scans at 12 months of age on the participants and in analyzing these scans and others already collected. |
Dean Felsher https://profiles.stanford.edu/dean-felsher | Medicine-Oncology | My laboratory studies how oncogenes initiate and maintain tumorigenesis. We have discovered that cancers are "oncogene addicted" and defined both tumor intrinsic and host immune dependent mechanisms. In particular, we study how the the MYC oncogene pathway causes human cancer. We have developed novel approaches using conditional transgenic models to target the MYC pathway for the treatment of cancer using immune oncology, metabolic, microbiome and nanotechnology. The NIH has chosen my work to be funded by the NIH Outstanding Investigator Award. |
Liang Feng https://profiles.stanford.edu/liang-feng | Molecular and Cellular Physiology | Our research interest lies primarily in understanding the mechanism and regulation of membrane proteins, and developing their modulators for potential therapeutic and biotechnological applications. We are taking a multi-disciplinary approach, employing structural methods to capture the high-resolution picture of different states, functional assays and biophysical methods to dissect the information inferred from the structure and computational approaches to understand their dynamics and kinetics. |
Marcelo Anibal Fernandez-Vina https://cap.stanford.edu/profiles/frdActionServlet?choiceId=printerprofile&profileversion=full&profileId=21099 | Pathology | I have expertise in the fields of Histocompatibility and Immunogenetics. My research interests are focused in identifying HLA mismatches that may be better tolerated than average and identify the molecular basis for such tolerance. My research has also focus in the identification of HLA encoded factors that associate with both, susceptibility and resistance to development of diseases as identifying HLA factors that may dictate disease course post infection. |
Katherine Ferrara https://profiles.stanford.edu/katherine-ferrara | Radiology | Molecular imaging, drug delivery, personalized medicine. Ultrasound, MRI, PET, sequencing, nanoparticles. |
Polly Fordyce https://profiles.stanford.edu/polly-fordyce | Bioengineering, Genetics | Microfluidics for high-throughput biophysics, biochemistry, & single-cell biology; protein engineering |
Joseph D Forrester https://profiles.stanford.edu/joseph-forrester | School of Medicine, Department of Surgery, Division of General Surgery, Section of Acute Care Surgery | The Forrester Lab has two primary research themes: 1) Finding better ways to predict the distribution of healthcare-associated infections through smart infection surveillance in high- and low-resource settings 2) Improving the operative care of patients with chest wall injury To address these topics, we use a combination of techniques, drawing from field epidemiology, biostatistics, and computer science. Dr. Forrester leverages available resources in the Section of Acute Care Surgery including the National Trauma Databank, Trauma Quality Improvement Program, the Healthcare Quality Improvement Database series, and the PearlDiver databases – among other clinical databases. Dr. Forrester oversees and mentors a group of undergraduates, medical students, surgical residents, and critical care fellows with whom the post-graduate researcher would work. |
Paige M Fox https://profiles.stanford.edu/paige-fox | Surgery | I am interested in improving patient care through tissue engineering and technology. My lab has multiple projects on a wide spectrum of conditions. We examine optimizing wound healing of chronically infected wounds through the delivery of local antibiotics in a collagen rich hydrogel. We use a model of chronic nerve compression to study and optimize muscle recovery after nerve release. We also examine enthesis healing after chronic injury. My lab collaborates with the School of Engineering to develop wireless microsensors to detect changes in blood flow. |
Richard Frock https://profiles.stanford.edu/richard-frock | Radiation Oncology | Overview: Our research is centered on understanding the fundamental and coordinate processes that repair our genome when double-stranded DNA ends become exposed. We employ a multi-functional DNA junction mapping platform designed to infer the genome-wide impacts of modulating DNA integrity. Our technology of choice, the high-throughput genome-wide translocation sequencing (HTGTS) platform, as a core function identifies recurrent “prey” double-stranded DNA breaks (DSBs) via their translocation to a “bait” DSB. We leverage this junction mapping platform in a number of different contexts to gain a better understanding of nuclear processes. Our functional genomics projects cover many different fields: radiation and cancer biology, genome editing, immunology, and cell cycle phase restricted DSB repair. |
Margaret Fuller https://med.stanford.edu/profiles/margaret-fuller#bio | Developmental Biology | We study the regulation of self-renewal, proliferation and differentiation in adult stem cell lineages using spermatogenesis in the fruitfully Drosophila as a model. Molecular mechanisms that trigger the switch from proliferating precursor cells to onset of differentiation start with a cascade of RNA binding proteins, lead through alternative processing of nascent mRNAs, to translational control, to novel mechanisms of transcript initiation. |
Lawrence Fung https://profiles.stanford.edu/lawrence-fung | Psychiatry & Behavioral Sciences | Dr. Lawrence Fung is a scientist and psychiatrist specialized in autism spectrum disorder (ASD), and the father of a neurodiverse teenager with ASD. He is the director of the Stanford Neurodiversity Project, which strives to uncover the strengths of neurodiverse individuals and utilize their talents to increase innovation and productivity of the society as a whole. He directs the Neurodiverse Student Support Program, Neurodiversity at Work Program, and Adult Neurodevelopment Clinic at Stanford. Dr. Fung is an assistant professor in the Department of Psychiatry and Behavioral Sciences at Stanford University. His lab advances the understanding of neural bases of human socio-communicative and cognitive functions by using novel neuroimaging and technologies. His team devise and implement novel interventions to improve the lives of neurodiverse individuals by maximizing their potential and productivity. For example, he is conducting a study to demonstrate that specialized employment programs such as Neurodiversity at Work program will result in higher retention rates and quality of life. |
Pascal Geldsetzer https://profiles.stanford.edu/pascal-geldsetzer | Assistant Professor (University Tenure Line) in the Division of Primary Care and Population Health, Department of Medicine, Stanford School of Medicine | Research interests: Application of novel causal inference techniques in electronic health record data to study the effectiveness of various clinical interventions; development of machine learning tools and innovations to improve healthcare in low- and middle-income countries (LMICs); deep learning in satellite imagery to monitor health in LMICs; epidemiology of chronic conditions and infectious diseases in LMICs; health effects of air pollution; climate change and health; health services research and impact evaluations in LMICs; clinical research; bioinformatics; environmental health; epidemiology; decision sciences; health policy; global health; population health Below is an example list of projects. Project 1: Regression discontinuity in electronic health record data to individualize care Randomized clinical trials (RCTs), which in clinical medicine are widely believed to be the only study design that can reliably establish causal effects, have important drawbacks. These disadvantages include that they are costly and time-consuming, study participants are often not representative of “real-word” patients, interventions tend to be more closely supervised than in routine care, outcomes are frequently short-term surrogate measures for longer-term health outcomes, and they are usually not powered to examine how treatment effects vary across granular patient subgroups. Although observational studies of large-scale electronic health record (EHR) data could overcome these weaknesses, such studies are thought to yield unreliable causal effect estimates because they assume that all possible confounders were perfectly measured and modeled in the analysis. This project aims to establish a study design for EHR data – regression discontinuity (RD) – that is widely used in the social sciences but rarely in clinical medicine and epidemiology. RD can be used to ascertain the causal effect of an intervention whenever there is a discrete threshold in a continuous variable that affects the probability of receiving the intervention. Such thresholds are ubiquitous in clinical medicine. For instance, decisions on diagnosing diabetes (blood glucose/HbA1c threshold), providing a blood transfusion (hemoglobin threshold), and prescribing antihypertensive medications (blood pressure threshold) are all at least partially driven by thresholds in continuous variables. Intuitively, RD mimics a randomized trial because in clinical medicine the continuous variable on which the threshold is based is usually measured with at least some degree of random measurement error. Thus, if the analysis “zooms in” around the threshold, patients will, in effect, have been randomized to being just above or just below the threshold. We have numerous ongoing analyses in a variety of different areas of medicine and focussed on a variety of interventions, including medications, lifestyle advice, and different care pathways. Students are also encouraged to bring their own analysis ideas to which this method can be applied. Requirements: strong quantitative background and experience coding in R or another statistical software. Project 2: Applying deep learning to satellite imagery to obtain real-time estimates of health indicators for each village and neighborhood in a country Globally, child and maternal health has improved substantially in the 21st century: thenumber of children dying before reaching age five has more than halved even in some ofthe world’s poorest regions. Despite the global progress, in some regions more than halfof young children grow up malnourished and one in five fail to reach their fifth birthday.These regions are also data deserts. Future progress on health in the world’s most underserved regions requires better understanding of the location, extent, distribution, and changes in health and disease. To date, the most important and extensive effort to quantify the health burden in thedeveloping world is the Global Burden of Disease project. The Global Burden of Disease relies on sparse, costly, and infrequent surveys and studies to estimate basic quantities such as mortality or malnutrition. We propose to change the paradigm by using directly observed information to infer several basic health metrics in the developing world: female body mass index, child malnutrition, maternal health, and child mortality. Using high spatial and temporal resolution remote sensing information, including daytime imagery, climate information, nightlights, and land use data, we propose to create tools for directly monitoring the health of populations across the developing world. Remote sensing information has been used to predict water quality, roads and connectivity to services, crop yields, droughts and growing conditions, infrastructure (especially water pipes and electrification), and economic activity, all of which have known bearing on health. This project is being conducted in close collaboration with Profs. Eran Bendavid and Stefano Ermon. Requirements: experience or keen interest in convolutional neural networks for image-based data. Project 3: Retinal fundus photographs to predict diabetes: A potential solution for community-based diabetes screening in low- and middle-income countries Community health workers (CHWs) in low- and middle-income countries (LMICs) are often not allowed to handle blood, excluding the possibility of CHW-led diabetes screening using blood glucose testing. Even in settings in which CHW-led blood testing is a possibility, screening campaigns frequently rely on a random rather than a fasting blood glucose test because of logistical difficulties in ensuring that those who are screened are fasted. Random blood glucose as well as questionnaire-based methods for screening for diabetes perform poorly. The fact that it is becoming increasingly common for CHWs to be equipped with smartphones or tablets to assist them with their work may open up new possibilities for diabetes screening in LMICs. Smartphones can already be used to take photos of the retinal fundus, and it is likely that there will be an increasing number of technological tools to take high-quality retinal fundus images in inexpensive ways in the future. Concurrently, convolutional neural networks (CNNs) are increasingly being applied successfully in medical imagery. This includes retinal fundus photographs, for which CNNs have been used to predict diabetic retinopathy (a highly prevalent condition among adults with diabetes in LMICs due to poor rates of diabetes control) among patients with diabetes, as well as age, gender, and smoking status. In order to investigate whether retinal fundus photography via smartphones could be a promising method to screen for diabetes in LMICs, this study aims to determine how reliably convolutional neural networks in retinal fundus photographs taken in the UK Biobank cohort predict a person’s diabetes status. Requirements: experience or keen interest in convolutional neural networks for image-based data. Project 4: Predicting cardiovascular disease risk and mortality using deep learning in retinal fundus images Retinal fundus images are relatively easy to generate, in the sense that the necessary process is non-invasive and could potentially be accomplished with a smartphone camera. Retinal fundus images have already been used in the context of image-based machine learning mostly for diagnosing diabetic retinopathy. Since pathological changes in the retina can be the result of high blood pressure, changes in the microvasculature due to diabetes, or potentially effects from other cardiovascular disease risk factors, we want to investigate to what degree cardiovascular disease risk and all-cause mortality can be predicted using an image-based machine learning model. The addition of deep learning in retinal images to cardiovascular disease risk equations might improve the predictive ability of cardiovascular disease risk equations that are currently used in clinical practice. Our research question is whether and to what extent deep learning in retinal fundus images adds to the predictive accuracy of existing risk scores for cardiovascular disease events. The motivation of this work is to ascertain whether retinal fundus images should be added to cardiovascular disease risk scores or could even replace them in settings in which doing a full cardiovascular disease risk assessment is difficult (e.g., low-income countries with restricted access to a laboratory). The aims of this analysis are to i) compare the predictive accuracy of existing cardiovascular disease risk scores with those from a convolutional neural network in retinal fundus images, and ii) given the large proportion of deaths that are caused by cardiovascular disease, determine how well deep learning in retinal fundus images predicts all-cause mortality. In the long run, should we be able to show that the analysis of retinal images provides reliable information on individuals' cardiovascular disease risk and/or all-cause mortality, such an analysis might become a non-invasive, quick, and cost-efficient risk stratification tool for clinical care. The data source for this project is the UK Biobank. Requirements: experience or keen interest in convolutional neural networks for image-based data. Project 5: How has health-related behavior and knowledge changed over time in low- and middle-income countries? We have achieved great success in global health with improving coverage of certain healthcare products. For example, coverage of antiretroviral therapy has gone up enormously and vaccination coverage is also (albeit not perfect) quite high in most countries, even the poorest. The hypothesis of this project is that global health efforts aimed at improving knowledge and bringing about behavior change have had very little impact, despite huge amounts of money having been spent on these programs. This project aims to describe how indicators of knowledge and health-related behavior have changed over time. We will use all questions on either knowledge or behavior that are in the Demographic and Health Surveys or the UNICEF Multiple Indicator Cluster Surveys. Specifically, we will analyze all questions that have been asked in at least two survey rounds in the same country. Large parts of the required dataset have already been assembled by our team. Requirements: experience coding in R or another statistical software. Project 6: What is the effect of air pollution on maternal and child health outcomes? Determining the health effects of air pollution has been difficult due to the multitude of potential confounding variables in the relationship between air pollution and health. This project aims to exploit a natural experiment to estimate the causal effect of air pollution on maternal and child health outcomes. Air pollution in many areas of South Asia is seasonal, with large spikes in particulate matter concentration occurring in the winter months. Whether or not a pregnancy is exposed to this winter spike in air pollution is random if the timing of conception is unrelated to any factors that are linked to maternal and child health outcomes. Using this analytical strategy, we aim to compare pregnancies that were exposed to this winter spike in air pollution with those that were not. The data source for this project will be the Demographic and Health Surveys conducted in relevant regions of South Asia (particularly India, Bangladesh, and Nepal). A first step in the analysis will be to test to what degree the timing of conception is correlates with observable characteristics of mothers (e.g., household wealth and education). Requirements: experience coding in R or another statistical software. |
Andrew Gentles https://med.stanford.edu/profiles/andrew-gentles | Medicine (Biomedical Informatics) | Computational genomics and proteomics to understand biology of normal and disease. Much of our work focuses on cancer and (increasingly) neurodegenerative disease. |
Paul George https://profiles.stanford.edu/paul-george | Neurology and Neurological Sciences | Our lab evaluates neural recovery through the use of biological (ie stem cells) and bioengineering tools with the ultimate goal of understanding recovery mechanisms and translation to clinical therapies. |
Olivier Gevaert https://profiles.stanford.edu/olivier-gevaert | Dept. of Medicine | Vast amounts of molecular data characterizing the genome, epi-genome and transcriptome are becoming available for a wide range of complex disease such as cancer and neurodegenerative diseases. In addition, new computational tools for quantitatively analyzing medical and pathological images are creating new types of phenotypic data. Now we have the opportunity to integrate the data at molecular, cellular and tissue scale to create a more comprehensive view of key biological processes underlying complex diseases. Moreover, this integration can have profound contributions toward predicting diagnosis and treatment. The Gevaert lab focuses on achieving progress in multi-scale modeling by tackling challenges in biomedical multi-scale data fusion. Applications are in the area of complex diseases with most projects in the lab focused on oncology, and possible new directions studying neuro-degenerative & cardiovascular diseases. |
William Giardino https://profiles.stanford.edu/william-giardino | Assistant Professor, Department of Psychiatry and Behavioral Sciences | The Giardino Laboratory of Circuits & Systems Neuroscience is a multidisciplinary team of research scientists that aim to decipher the neural mechanisms underlying psychiatric conditions of stress, addiction, and sleep disturbances. Our work uses genetic, pharmacological, physiological, anatomical, and computational approaches in freely behaving mice to monitor, map, and manipulate the neural circuits that drive approach/avoidance behaviors, drug-seeking, food intake, social interactions, and sleep/wake arousal states. |
Aaron Gitler https://profiles.stanford.edu/aaron-gitler | Genetics | Mechanisms of neurodegeneration. Parkinson’s disease, ALS. |
Anna Gloyn https://profiles.stanford.edu/anna-gloyn | Pediatrics | Dr Gloyn's current research projects are focused on the translation of genetic association signals for type 2 diabetes and glycaemic traits into cellular and molecular mechanisms for beta-cell dysfunction and diabetes. Her group uses a variety of complementary approaches, including human genetics, functional genomics, physiology and islet-biology to dissect out the molecular mechanisms driving disease pathogenesis. Keywords: Human Genetics, Diabetes, Functional Genomics, CRISRP, Stem Cell Biology, Islet Cell function |
Garry Gold https://profiles.stanford.edu/garry-gold | Radiology | MRI, Imaging, Human Performance, Sports, Musculoskeletal Disease |
Jeffrey Louis Goldberg https://profiles.stanford.edu/jeffrey-goldberg | Ophthalmology | Our research lab is focused on fundamental neuroscience of neuronal survival and axon growth, relevant to neuroprotection and regeneration. We use molecular and animal models in retinal ganglion cells and the visual system to discover and study signaling pathways. We also maintain a clinical/translational research program, studying novel structural and functional biomarkers and candidate neuroprotection therapeutics in human patients with glaucoma and other optic neuropathies. |
Lisa Goldman Rosas https://profiles.stanford.edu/lisa-goldman-rosas | Epidemiology and Population Health, SoM | health equity, social determinants of health, community-based participatory research |
Lisa Goldman Rosas https://profiles.stanford.edu/lisa-goldman-rosas | Epidemiology and Population Health | Dr. Goldman Rosas and the Food for Health Equity Lab focus on generating evidence for community health centers, policy makers, healthcare payors, and community-based organizations to effectively address food and nutrition insecurity in a way that promotes health equity. This work particularly focuses on improving nutrition and reducing chronic diseases within under-resourced communities and communities of color. Currently, the Food for Health Equity Lab is multiple randomized controlled trials, evaluations, and pilot projects to examine the effectiveness of innovative "Food as Medicine". Each project involves the delivery of healthy foods to patients and behavior support that helps patients develop and maintain healthy habits. Dr. Goldman Rosas uses a community-based participatory research approach that features shared leadership with patients, caregivers, community partners, and policy makers. |
Natalia Gomez-Ospina https://profiles.stanford.edu/natalia-gomez-ospina | Pediatrics | The main focus of Dr. Gomez-Ospina’s lab is to develop therapies for patients with genetic diseases. The lab uses genome editing and stem cells to produce definitive treatments for childhood neurodegenerative diseases, many of which are lysosomal storage disorders. Current projects in the lab include developing autologous transplantation of genome-edited hematopoietic stem cells for Mucopolysaccharidosis type I, Gaucher, Krabbe disease, Frontotemporal Dementia, and Friedreich's ataxia. Although there is a strong translational focus to the lab, we are also pursuing basic science questions to understand and enhance our therapies including: 1) increasing the efficiency of genome editing tools, 2) understanding microglia turnover in response to conditioning before hematopoietic stem transplant, and 3) establishing brain-specific conditioning regimens. |
Miriam B. Goodman https://profiles.stanford.edu/miriam-goodman | Molecular and Cellular Physiology | The wormsense neuroscience lab combines genetic dissection with cellular physiology, quantitative behavioral studies, and cell biology to deconstruct the molecular basis of mechanical and chemical sensation. We use sensory transduction in C. elegans as a window into how we feel, how neurons bend without breaking, and how the failure to regenerate underpins disease- and drug-induced neuropathy. We also co-lead NeuroPlant, a multidisciplinary project that links expertise in plant systems biochemistry (Rhee) and nematode chemosensation (Goodman) to identify plant-derived compounds that modulate neurons. The group also maintains active collaborations with engineers and physicists to develop novel devices and tools for delivering and measuring force in the nervous system. |
Stuart Goodman https://profiles.stanford.edu/stuart-goodman | Orthopaedic Surgery and Bioengineering | Dr. Goodman's clinical research interests center on the outcome of surgery for arthritis including primary and revision total joint replacement, juvenile arthritis, and osteonecrosis of the hip and knee. His basic science interests center on biocompatibility of orthopaedic implants, the innate immune system, inflammation, osteonecrosis, and musculoskeletal tissue regeneration and repair. We perform in vitro and in vivo studies and use cell and molecular biology techniques, RNA-seq, CyTOF, micro CT, and other methods of analysis. |
Edward Graves https://profiles.stanford.edu/edward-graves | Radiation Oncology | My research group is focused on applications of emerging functional and molecular imaging techniques in radiation therapy of cancer. In order to integrate these novel imaging procedures with state-of-the-art radiation therapy, a number of issues must be addressed. First, what are the molecular targets that hold the most promise for targeting and monitoring response to radiation therapy, and how can they best be visualized in vivo? Second, what are the limitations of novel imaging techniques that may bear on their application in radiation oncology? Third, how can one display, analyze, and segment multiple three-dimensional datasets to generate target volumes for radiotherapy? And finally, how will the information contained in imaging results of different modalities be integrated into the selection of a treatment course for a patient and subsequently, where appropriate, the specification of an optimized radiation target? These questions comprise my research. Projects that address these topics include the implementation and evaluation of clinical PET/CT imaging for radiation treatment planning, development and validation of novel molecular imaging methods for preclinical and clinical imaging of tumor radiosensitivity and radiation response, development of software for multimodal image analysis, and study of tumor hypoxia and radioresistance in small animal models using a multimodality molecular imaging approach. |
William Greenleaf https://profiles.stanford.edu/william-greenleaf | Genetics | Single-cell methods, genomics, genetics, epigenetics, high-throughput methods, biochemistry, biophysics, sequencing, cancer, developmental biology. |
Michael D. Greicius https://profiles.stanford.edu/michael-greicius | Neurology and Neurological Sciences | My lab uses imaging and genetics to understand the biological underpinnings of Alzheimer's disease and related disorders across scales from molecule to cell to network to behavior. |
Nicolas Grillet https://profiles.stanford.edu/nicolas-grillet | Assistant Professor in School of Medicine Otolaryngology department | "How do we hear?" and "why are we all becoming deaf" are the central questions that we are tackling in our lab. We are particularly interested in the sensory cells of the inner called the hair cells, capable of converting a mechanical force induced by sound into an electric output. This conversion, called mechanotransduction, occurs in an elaborated apical structure of the hair cell made of membrane protrusions interconnected by filaments. The filaments pull on the membrane and open mechanosensitive ion channels. We have identified by genetics means a new component of this machinery that regulates the channel's activity. Mutation in this gene cause hearing loss in human, dogs, and mice. The particular questions that we want to answer at the moment are: - Can overexpression of the gene rescue the mechanotransduction defect, and which domains of the protein are necessary for it? How? using overexpression in the dissected mouse inner ear epithelium from a mutant mouse, cultured and electroporated with a cDNA; after one day of culture, the hair cells are mechanically stimulated by a fluid-jet, and the mechanotransduction response is monitored with a genetically encoded calcium sensor. - Where is the protein localized within the apical specialization of hair cells? How? We have genetically tagged this gene in the mouse, enabling us to detect the protein with an antibody. Here we want high-resolution localization, so we will use transmission electron microscopy and secondary antibodies conjugated with a gold bead, stopping the electron beam. We will do the observation of the sections by Transmission Electron Microscopy. We will prepare samples to fix them optimally for structure preservation to have the imaging facility generating sections and immunostaining for us. |
Tanja Andrea Gruber https://profiles.stanford.edu/tanja-gruber | Department of Pediatrics | Pediatric high risk acute leukemia including AML and KMT2Ar ALL. Genomics, high throughput drug screening, and murine modeling. |
Jennifer Hah https://profiles.stanford.edu/Jennifer_Hah | Anesthesiology | I study the link between mood and opioid use with a focus on persistent opioid use after surgery. We study novel interventions to promote opioid cessation. We conduct extended longitudinal data collection and interface with bioinformatics for machine learning and NLP chart review. |
Summer Han https://profiles.stanford.edu/summer-han | Neurosurgery and Medicine | Developing machine learning based prediction models for cancer risk and outcomes |
Brian Hargreaves https://profiles.stanford.edu/brian-hargreaves | Radiology | Medical Imaging, Magnetic Resonance Imaging, Mixed-Reality in Medicine. Applications to body, breast and musculoskeletal diseases. |
Keren Haroush https://profiles.stanford.edu/keren-haroush | Neurobiology | We study the neuronal basis of dynamic social interactions. We perform cutting-edge large-scale recordings and targeted perturbation in rich social tasks based on game-theory combined with advanced analytical approaches, brain-computer interface, machine vision and deep learning to understand the single neuron and population level underpinnings of complex social computations, such as cooperation, empathy, fairness and reciprocity. |
Melanie Hayden Gephart https://profiles.stanford.edu/MHaydenGephart?releaseVersion=7.23.0 | Neurosurgery | Our laboratory accelerates translational brain tumor research, combining innovative techniques in genetics and cancer biology with a unique insight into the pressing clinical questions facing patients. We use novel genetic sequencing methods and modeling to understand how cancer grows in the brain, inadvertently supported by native brain cells. We focus on translational therapeutic targets identified from and validated with primary human tissue and cerebrospinal fluid whenever possible, as this most reliably reflects the human disease. These findings have led to novel diagnostic tools and clinical trials for patients with malignant brain tumors. Our laboratory is a dynamic and collaborative working environment, benefitting from the supportive research environment at Stanford. Our laboratory space bridges the Stanford core campus and the medical facilities, emblematic of the translational aspects of our work. |
Jill Helms https://profiles.stanford.edu/jill-helms | Surgery | I trained as a dentist and have a certificate in Periodontics and a PhD in Developmental Neurosciences. My laboratory works in the field of Regenerative Medicine and Dental Medicine. We focus on the regulation of tissue repair and regeneration with an eye towards understanding how basic science discoveries can be translated into therapeutic interventions that improve patient outcomes. In addition to being a Professor, I am also the co-founder of Ankasa Regenerative Therapeutics, a venture-backed biopharmaceutical start-up in South San Francisco. While conducting clinically relevant research has been my main objective, it goes hand-in-hand with another goal: I firmly believed that education is one of the most important tools to improving human health, and I aims to use every avenue available to transform the way people think about science and medicine, and emphasize its contribution to their daily lives. |
Tina Hernandez-Boussard Associate Professor | Medicine, Biomedical Informatics | My background and expertise are in the field of biomedical informatics, health services research, and epidemiology. My current work utilizes high-volume digital data to accurately and efficiently monitor, measure, and predict healthcare outcomes using natural language processing and machine/deep learning techniques. More recently, my work focuses on the development of artificial intelligence technologies to help deliver precise, fair and patient-centric treatment approaches that maximize benefits and minimize risks for patients. |
Daniel Herschlag https://profiles.stanford.edu/daniel-herschlag | Biochemistry | Understanding biology and its evolution from the standpoint the atomic and molecular forces that guide and constrain life and its evolution, with an emphasis on RNA, RNA binding proteins, and enzymes and their catalysis and evolution. Pioneering high-throughput quantitative studies of RNA and proteins and Quantitative Cellular Biochemistry. |
Hadi Hosseini https://profiles.stanford.edu/seyed-mohammad-hadi-hosseini | Stanford Psychiatry | Developing advanced computational and engineering approaches to decode neural network signatures of mental illnesses: We leverage multimodal neuroimaging (MRI, NIRS), advanced network science and machine learning techniques to investigate alterations in the organization of structural and functional brain networks in various neuropsychiatric, neurodegenerative and neurodevelopmental disorders. We also develop and test novel, brain-focused cognitive interventions for targted enhancement of memory and executive functioning given their involvement in a variety of brain disorders. Undergoing studies in my lab include: - Identifying connectome-markers of brain disorders. - Advanced computational approaches for integrating multimodal data to capture heterogeneity of brain disorders - Neuromonitoring guided cognitive augmentation for children with ADHD. - Multimodal neuroimaging study to examine neuroplasticity in older adults at risk of developing Alzheimer’s disease. - Developing low-cost, wearable, optical imaging system for at-home functional neuroimaging and neuro-intervention |
Michael Howitt Assistant Professor | Pathology | My lab studies the interactions between the microbiota and the immune system in the gut. We are particularly interested in understanding how the sense of taste in extra-oral tissues monitors microbes and communicates with the immune and nervous systems. We employ mouse models, primary organoids, and computational approaches to answer these questions. |
Yang Hu https://profiles.stanford.edu/yang-hu | Ophthalmology | The ultimate goal of the laboratory is to develop efficient therapeutic strategies to achieve CNS neural repair, through promoting neuroprotection, axon regeneration and functional recovery. Specifically, we are using state-of-the-art tools and creating novel tools to study retinal ganglion cell (RGC) and optic nerve in various optic neuropathies including traumatic, glaucomatous and inflammatory optic nerve injuries, to fully understand the molecular mechanisms of CNS neurodegeneration and axon regeneration failure. |
Ngan Huang https://med.stanford.edu/huanglab.html | Cardiothoracic Surgery | cardiovascular tissue engineering, biomaterials, stem cells, 3D printing |
John Huguenard https://huguenardlab.stanford.edu | Neurology and Neurological Sciences | Neuroscience, epilepsy, real-time detection and treatment of seizures, electrophysiology, imaging, behavior, optogenetics |
Juliana Idoyaga https://profiles.stanford.edu/juliana-idoyaga | Microbiology and Immunology Department | The Idoyaga Lab is focused on the function and biology of very unique cells of the immune system, Dendritic cells (DCs). DCs are specialized antigen-presenting cells that initiate and modulate our body’s immune responses to invading microbes. DCs also play a crucial role in maintaining immune unresponsiveness to our own tissues and environmental and/or innocuous substances. Taking advantage of high-dimensional approaches such as CyTOF, scRNA-seq and ATAC-seq, our goal is to dissect the basic biology of DCs in health and disease with the goal of designing novel treatments. |
Prasanna Jagannathan https://profiles.stanford.edu/prasanna-jagannathan | Medicine Infectious Diseases | Dr. Prasanna Jagannathan is an Infectious Diseases specialist with a research program in human immunology and clinical trials of immune modulatory interventions. His goals are to further our understanding of the mechanisms of naturally acquired immunity to malaria and, more recently, SARS-CoV-2. These studies bridge epidemiologic studies and clinical trials, cellular immune profiling techniques including multiparameter flow cytometry, transcriptomics, and epigenetics, and in vitro mechanistic studies. He is leading a study using systems biology approaches to study naturally acquired immunity to malaria in Ugandan children, and is soon beginning a clinical trial in Uganda that will test the impact of antimalarial chemoprevention on immune development in Ugandan infants. Dr. Jagannatahn has also been testing novel therapeutics for the treatment of COVID-19. Along with Dr. Upi Singh at Stanford, Dr. Jagannathan led a randomized, placebo-controlled trial of Peginterferon lambda for patients with mild to moderate symptoms, and his lab is studying host immune responses in these individuals. |
Michelle James https://profiles.stanford.edu/michelle-james | Assistant Professor Dept of Radiology | The primary aim of the James Lab is to improve the diagnosis and treatment of brain diseases by developing translational molecular imaging agents for visualizing neuroimmune interactions underlying conditions such as Alzheimer’s disease, multiple sclerosis, Myalgic Encephalomyelitis/Chronic Fatigue Syndrome, and stroke. We are researching how the brain and its resident immune cells interact with the peripheral immune system at very early, through to late, stages of disease. Our approach involves the discovery and characterization of clinically relevant immune cell biomarkers, followed by the design of imaging agents specifically targeting these biomarkers. After preclinical validation, we translate promising imaging probes to the clinic to enable precision targeting of immunomodulatory therapeutics and real-time monitoring of treatment response. The main technologies and/or techniques we employ are: Radiochemistry, neuroimaging of rodents and humans using positron emission tomography (PET imaging), CT, and MRI. |
James Kahn Professor | Medicine | Narrative medicine and clinical research |
Ioannis Karakikes https://profiles.stanford.edu/ioannis-karakikes | School of Medicine - Cardiothoracic Surgery | Our research is focused on the identification disease-specific mechanisms of genetic cardiomyopathies, such as dilated cardiomyopathy (DCM), and develop mechanism-based therapeutics. We employ an interdisciplinary approach, integrating functional genomics approaches in human induced pluripotent stem cell (hiPSC)-derived cardiovascular cells with single-cell transcriptomics and epigenetics to study cardiomyopathies in a genetically controlled and systematic manner. We have used this platform to uncover novel mechanisms underlying the pathogenesis of LMNA- (Lee et al, Nature, 2019) and PLN-related DCM (Feyen et al., Circulation, 2021). |
Mark Kay https://profiles.stanford.edu/mark-kay | Pediatrics | Gene Therapy and Genome Editing – Vector design, vector screens in whole mammals, epigenomic state of vector DNA in tissues, vector-mediated homologous recombination and treatment in animal models of human disease Non-coding RNA biology- Functional studies of various non-coding RNAs including: tRNA-derived small RNAs, Line 1 RNAs, and long non-coding RNAs and their processed miRNAs. |
Corey Keller https://profiles.stanford.edu/corey-keller | Assistant Professor, MCL, Psychiatry and Behavioral Sciences | The overarching goal of Dr. Keller’s Laboratory (kellerlab.stanford.edu) is to improve brain stimulation treatment for neurological and psychiatric disease. Specifically, his lab seeks to improve Transcranial Magnetic Stimulation (TMS) and other brain stimulation techniques by better understanding the fundamental principles of human brain plasticity and building trans-diagnostic real-time monitoring platforms for personalized brain stimulation. We use an array of neuroscience methods to better understand the basic principles of how to create change in brain circuits. We use this knowledge to develop more effective treatment strategies for depression and other psychiatric disorders. |
Saad Khan https://profiles.stanford.edu/saadkhan | Assistant Professor | Cancer>translational research>bench to bedside back to bench research analysis. My research is specifically focused on improving cancer outcomes, using data analytic techniques as well as hands-on experiments in cell lines/animals and patients. An example would be a patient with cancer on hospice who ingested homemade silver nanoparticles and had his cancer completely disappear! We did a physical/chemical analysis of the tumor and are now conducting cell line/mice experiments to identify if silver nanoparticles have any anticancer effect and then how to mass produce it. https://onlinelibrary.wiley.com/doi/full/10.1002/hed.25492 This experience would be best suited for those scientists in training who are considering applying to MD programs, or want to demonstrate research that actually impacted patients. I have 9 years of experience in mentoring students into MD programs, short courses, residencies and faculty. Deliverables to the scholar within the first 2 years: 1. learn translational cancer research techniques 2. 2-5 abstracts presented at national conferences 3. 2-3 manuscripts as first author The experience will be tailored to the individual scholar's skillset/interest. Probably best suited for those who want to develop their experimental design experience and learn how to analyze/present data. |
Mathew Kiang https://profiles.stanford.edu/mkiang | Epidemiology and Population Health | Primarily focused on social epidemiology and computational epidemiology. Current projects include understanding racial/ethnic disparities in drug poisoning deaths (especially opioids) and treatment for substance use disorder, understanding socioeconomic and medical vulnerability in the context of power outages and wildfires, and using smartphone-based digital phenotyping data to get better measurements of human behavior. |
Peter S. Kim https://med.stanford.edu/profiles/peterkim | Biochemistry | We aim to create vaccines for important infectious diseases. Our research is primarily focused on antibody-mediated immune responses. |
Seung Kim https://med.stanford.edu/profiles/seung-kim | Developmental Biology | Our group investigates pancreas and hormone biology related to metabolism, and human diseases like diabetes mellitus and pancreatic cancer. We use a wide range of approaches and experimental systems to address outstanding questions in our fields of research, including experimental animal models, human stem cells, and patient-derived tissue samples. |
Feliks Kogan https://profiles.stanford.edu/feliks-kogan | Radiology | My Group works on the development and clinical translation of novel imaging technologies geared toward studying musculoskeletal function and detection of musculoskeletal disease at the earliest stage. Our group is developing novel PET and MRI methods to study early and reversible tissue changes at the cellular and molecular level, functional imaging of joint mechanics, physiology and tissue microstructure, and rapid and comprehensive MRI methods for early, low-cost, and precise detection of musculoskeletal disease. |
Sheri Krams https://profiles.stanford.edu/sheri-krams | Surgery. and Immunology | Our research team investigates the response to viruses (SARS-CoV-2, EBV) and alloantigen with a focus on human immunology. The lab utilizes state of the art technologies to examine the underlying mechanisms of the innate and adaptive immune responses. |
Anshul Kundaje https://profiles.stanford.edu/anshul-kundaje | Genetics | Our primary research interests are computational biology and applied machine learning with a focus on gene regulation. My lab develops statistical and machine learning methods to infer integrative models of transcriptional regulation and interpret non-coding genetic variation through the lens of gene regulation. At the core of our research is the idea that obtaining a genome-wide and system-level understanding of gene regulation is essential to decipher the causal genetic and molecular basis of disease. What makes our research unique is the ability to condense massive compendia of genomic data into interpretable and predictive computational models capable of capturing functional heterogeneity and context-specificity thereby allowing the discovery of exceptions rather than focusing only on the norms. We collaborate extensively with experimental biologists within and outside Stanford to validate hypotheses generated by our models and discover novel biology. We have led the analysis efforts of the Encyclopedia of DNA Elements (ENCODE) and The Roadmap Epigenomics Projects. We are currently working on the following key research areas. (1) Interpretable deep learning models of protein-DNA binding, chromatin accessibility and chromatin state (2) Interpretable deep learning models of protein-RNA binding, RNA structure, splicing (3) Learning transcriptional regulatory networks that integrate cis-regulatory domains and activity of trans-regulators (4) Learning regulatory dynamics by integrating multi-omic single cell and bulk data from temporal (e.g. differentiation/reprogramming) and large-scale perturbation (e.g. CRISPR knockout/knockdown) experiments (5) Prioritizing and interpreting functional genetic variation and its impact on chromatin, expression and disease phenotypes (neurodegeneration, neuropsychiatric, T2D, cardiovascular disease, colorectal cancer) (6) Deconvolving cell-free DNA signals in cancer and pregnancy disorders |
Curt Langlotz https://profiles.stanford.edu/curtis-langlotz | Professor of Radiology | machine learning for natural language processing and computer vision |
Marvin Langston https://med.stanford.edu/profiles/marvin-langston | Epidemiology & Population Health | My lab is primarily focused on the epidemiology of urological conditions. We use use a variety of approaches and data from large national cohorts and electronic health records to consider projects related to early detection and screening, etiology of urological cancers, and inequities in cancer care and survival. Particularly we are interested in describing and mitigating inequities in cancer for racial and ethnic minoritized groups and sexual and gender minorities. |
Henry Lee https://profiles.stanford.edu/henry-lee | Pediatrics | Perinatal / neonatal epidemiology, health services research, and disparities |
Craig Levin https://profiles.stanford.edu/craig-levin | Professor Radiology | In our lab we explore new instrumentation and computational algorithms for non-invasively visualizing and quantifying the molecular basis of disease (e.g. cancer, neurological disorders) in living subjects using measurements made from outside the body. |
Ron Levy levy@stanford.edu | Medicine | Cancer and the Immune System |
Ronglih Liao https://profiles.stanford.edu/ronglih-liao ;https://med.stanford.edu/liaocmrl.html | CVMed | The scientific mission of Dr. Liao’s laboratory has been to elucidate the key mechanisms underlying the repair and regeneration of heart after myocardial injury. This has involved using functional genomics and proteomics approaches to investigate genes of interest in stem cells, isolated primary cardiac cells, organ and whole animals. In addition to her scientific pursuits, Dr. Liao has been fully invested in mentoring/promoting the next generation of investigators including under-represented and female scientists and in disseminating scientific knowledge to the larger research community. |
Michael Lim https://profiles.stanford.edu/238246 | Neurosurgery | My primary interest is in the immune microenvironment of Central Nervous System Cancers. I have 3 facets to my research: 1. Basic - we have been studying mechanisms behind immune cell tolerance and global immunosuppression in GBM. We are doing studies in both mice and human. 2. Translation - we have built an infrastructure to test novel therapeutics in mice for the purposes of translating them to clinical trials. We also have specimens from clinical trials that we are doing correlative studies. 3. Clinical trials - We have been able to take multiple agents from my laboratory and translate them to phase 1, 2, and 3 trials. |
Michael Lin https://profiles.stanford.edu/michael-lin | Neurobiology and Bioengineering | The Lin Lab applies protein design to investigate biology and treat disease. Topics include fluorescent reporting of neuronal electricity, luminescent reporting of cellular biochemistry, and rewiring of oncogenic signals to activation of therapeutic viruses. |
Jonathan Lin https://profiles.stanford.edu/216141 | Pathology | Retinal Cell Biology, Neurodegeneration, and Eye Pathology |
Michelle Lin https://profiles.stanford.edu/mplin | Emergency Medicine | Dr. Lin's active NIH-funded research portfolio includes developing a novel patient-reported outcome measure for emergency asthma care; evaluating post-acute transitions and outcomes for high-risk populations using administrative claims and mixed methods approaches; and enhancing health professions workforce diversity and retention. Her prior funded projects have evaluated the impact of value-based care on emergency care delivery and payment; drivers of ED admission rates; and changes in the intensity of emergency care. |
Eleni Linos PCRT.Stanford.edu | Dermatology | Public health, Epidemiology, Dermatology, Technology, Geriatrics |
Eleni Linos https://profiles.stanford.edu/eleni-linos | Dermatology | Public Health, Dermatology, Technology, Geriatrics, Diversity Equity and Inclusion, Health Disparities, Skin Cancer |
Wu Liu https://profiles.stanford.edu/wu-liu | Radiation Physics - Radiation Oncology | Theranostic nanoparticles for radiosensitization and medical imaging. Novel treatment and computation techniques. |
Billy W Loo http://profiles.stanford.edu/Billy_Loo | Radiation Oncology | Radiation oncology, radiation biology, medical physics, bioengineering |
Karl Lorenz https://profiles.stanford.edu/karl-lorenz | Primary care population health - palliative care | Quality measures, pain, symptoms, models of care, goals of care communication, cancer, palliative care, serious illness |
Stephen Luby https://profiles.stanford.edu/stephen-luby | Infectious Diseases and Geographic Medicine | Identifying and interrupting pathways of infectious disease transmission in low income countries. |
Bruce MacIver https://profiles.stanford.edu/m-maciver | Anesthesia | Neuronal Mechanisms for Loss and Recovery of Consciousness. |
Sean Mackey https://profiles.stanford.edu/sean-mackey | Division of Pain Medicine, Department of Anesthesiology, Periperative and Pain Medicine | Human pain research (neurosciences, outcomes, biomarkers, learning health systems). We use a broad range of technologies from neuroimaging, sensory testing, patient/clinician outcomes, multiomics, informatics). |
Merritt Maduke https://profiles.stanford.edu/merritt-maduke | Molecular & Cellular Physiology | Molecular mechanisms of ion channels and transporters; mechanisms of ultrasound neuromodulation |
Holden Maecker https://profiles.stanford.edu/holden-maecker | Microbiology & Immunology | A major aim of our lab is to define metrics of immune competence in various settings, including cancer immunotherapy, organ transplantation, allergy, and chronic viral infection. We use CyTOF mass cytometry, often in combination with other technologies, to broadly survey immune features at the cellular level, then examine links between features or groups of features and clinical outcome. A long-term goal is to create an assay of global immune competence that could predict risk for various immune-related outcomes in both healthy individuals and in disease. |
Vinit Mahajan https://profiles.stanford.edu/intranet/vinit-mahajan | Associate Professor of Ophthalmology | Dr. Mahajan's research team discovered the first gene to cause non syndromic uveitis and is now using protein crystallography to design therapeutic inhibitors for calpain-5. Mahajan and his team performed the first CRISPR gene editing therapy for eye disease in human stem cells. They have also created in vivo models for diabetic retinopathy and uveitis. Using translational proteomics, Mahajan’s multidisciplinary team is developing new precision health approaches using molecular biomarkers to diagnose retinal disease, select personalized therapies, and decode the anatomic structures of the human eye. |
Ravi Majeti https://profiles.stanford.edu/ravindra-majeti | Medicine | AML, stem cell biology, pre-leukemia, cancer immunotherapy, genome editing |
Alison Marsden https://profiles.stanford.edu/alison-marsden | Pediatrics | Cardiovascular biomechanics, patient specific modeling of blood flow for surgical planning |
Olivia Martinez https://profiles.stanford.edu/olivia-martinez | PROFESSOR, DEPT OF SURGERY | The Martinez lab has three major areas of study: 1) the immune response to viruses (EBV, SARS-CoV-2, and CMV); 2) pathogenesis of B cell lymphomas 3) immune pathways of transplant graft rejection. We approach all of these questions from the standpoint of understanding how T cells and NK cells recognize and respond to viruses, tumors or foreign tissue. The goal is to understand what determines whether an immune response to viruses and tumors is effective, and, conversely, how we can regulate the immune response to organ grants. We investigate these questions using specimens from the relevant patient groups, but also utilize animal models in combination with high dimensional approaches such as CyTOF, immune receptor sequencing, and other single cell assays of lymphoid cells. |
Mark McGovern https://profiles.stanford.edu/mark-mcgovern | Professor in Psychiatry and by Courtesy in Medicine | I am an implementation scientist. In our center, we advance methods and methodologies from this emerging discipline to improve access to the most effective treatments available, to enhance and ensure the quality of treatments being delivered, and to overcome health inequity and disparity. We work at the private and public health system level, on national, state, regional and local endeavors. Presently, our research is funded by NIH, DHHS, PCORI, SHC, VA, and private foundations. We are committed to advancing the quality of implementation science through mentorship, training and network development. |
Tracey McLaughlin Profiles.stanford.edu/Tracey-mclaughlin | Professor of Medicine, Division of Endocrinology | I conduct clinical/translational research related to obesity and insulin resistance/type 2 diabetes. Specifically, we perform deep metabolic phenotyping and then employ diet/surgical/medical weight perturbations (gain or loss) and perform adipose tissue biopsies to evaluate cellular and molecular characteristics as they relate to systemic insulin resistance/metabolic dysfunction. We are also exploring the potential for wearable devices (primarily continuous glucose monitoring) to augment current clinical approaches to identification of early stages of diabetes/metabolic dysfunction and precision diets. Opportunities for clinical and/or wet lab experience are available. |
Elizabeth Mellins https://profiles.stanford.edu/elizabeth-mellins | Professor of Pediatrics | My molecular immunology research group focuses on 2 main areas: (1) the role of class II major histocompatibiilty complex molecules (MHCII) in normal immunity and autoimmunity; (2) the role of MHCII-expressing monocytes in inflammation. Our approaches range from studying purified MHCII molecules in vitro, to cells expressing these molecules, including monocytes, to animal models. We use cutting-edge techniques including high dimensional cytometry (flow and CyTOF) and single cell RNA sequencing among others. |
Mark Mercola Professor | Medicine (CV med) | Cardiology, mechanisms of heart failure, induced pluripotent stem cell (iPSC), disease modelling, high throughput screening, therapeutic target discovery, drug discovery |
David Miklos https://profiles.stanford.edu/david-miklos | Medicine - BMT&Cell Therapy | Chimeric Antigen Receptor biology and other cell therapies Allogeneic immunity Graft versus host disease T and B cell interrogation using single cell RNA and immune receptor tracking |
Everett Moding https://profiles.stanford.edu/everett-moding | Assistant Professor of Radiation Oncology | Our laboratory performs translational cancer research by analyzing human tissue and blood samples with next-generation sequencing to understand the genetic underpinnings and expression signatures that determine treatment response and resistance. We then use preclinical models to validate our findings, perform mechanistic experiments, and test new therapies. |
Denise Monack https://profiles.stanford.edu/denise-monack | Microbiology and Immunology | We study genetic and molecular mechanisms of bacterial infections. Our projects range from mechanisms of intracellular pathogen survival/replication to characterizing interactions between bacterial pathogens and the microbiome within the guts of mammalian hosts. Most recently we are working to understand why specific Salmonella pathogens only infect humans - how did these microbes evolve? |
Elizabeth Mormino https://profiles.stanford.edu/elizabeth-mormino | Neurology and Neurological Sciences | Cognitive aging, Alzheimer’s disease, multimodality neuroimaging |
Anupama Narla https://profiles.stanford.edu/anupama-narla?tab=bio | Assistant Professor of Pediatrics (Hematology/Oncology) | Our lab studies pediatric bone marrow failures with a focus on understanding why mutations in ribosomes affect hematopoiesis and developing new drugs to stimulate the bone marrow with the ultimate goal of improving patient care. |
Aaron Newman https://profiles.stanford.edu/aaron-newman | Department of Biomedical Data Science (UTL) | Our group combines computational and experimental techniques to study the cellular organization of complex tissues, with a focus on determining the phenotypic diversity and clinical significance of tumor cell subsets. We have a particular interest in developing innovative data science tools that illuminate the cellular hierarchies and stromal elements that underlie tumor initiation, progression, and response to therapy. As part of this focus, we develop new algorithms to resolve cellular states and multicellular communities, tumor developmental hierarchies, and single-cell spatial relationships from genomic profiles of clinical biospecimens. Key results are further explored experimentally, both in our lab and through collaboration, with the goal of translating promising findings into the clinic. |
Mindie Nguyen https://med.stanford.edu/profiles/mindie-nguyen; | Medicine (GI Hepatology); By courtesy: Epidemiology and Population Health | My dry lab focuses on clinical outcome and epi population studies of patients with liver cancer, fatty liver, and viral hepatitis with focus on health disparity and linkage to care. Fatty liver affects about 30% of the world population. Hepatitis B affects about 290 million and viremic hepatitis C about 73 million people globally. Liver cancer is one of the cancers with highest mortality. We also conduct clinical trials and collaborate with other labs on lab based translational work. Over the last 15+ years, I have hosted several trainees from under-represented racial ethnic groups and from the most disadvantaged social economic groups. I myself came to the US at 17 as a refugee. I welcome and want to help trainees of all racial/ethnic, economic, social backgrounds. |
Teresa Nicolson https://profiles.stanford.edu/teresa-nicolson | OHNS | We study the molecular genetics of hearing and balance using zebrafish as our animal model. Many of the genes we have identified in screens for zebrafish mutants are associated with human hearing loss and vestibular dysfunction. Zebrafish are transparent and offer a number of advantages for studying the auditory/vestibular system. We use a wide variety of methods including behavioral assays, live cell imaging of transgenic lines, CRISPR editing, and RNAseq analysis to characterize mutants with peripheral and central defects. |
Daibhid O Maoileidigh profiles.stanford.edu/daibhid-o-maoileidigh | Otolarynglogy - Head & Neck Surgery | We are interested in how the ear converts mechanical stimuli into electrical signals that are transmitted to the brain. To investigate hearing and balance systems, we use mathematical, physics-based, and computational approaches. Our work sits at the intersection of many fields including physics, mathematics, mechanical and electrical engineering, neuroscience, and cellular biology. At present, we are focused on how the ear's sensory cells convert sound-induced stimuli into electrical currents and the neural response of the auditory system to sound. We collaborate closely with experimental labs to inform, validate, and test our computational models of hearing and balance systems. |
Lucy O'Brien https://profiles.stanford.edu/lucy-obrien | Molecular and Cellular Physiology | Our organs contain multitudes of cells—mature cells that execute organ function, stem cells that generate new cells, immature cells that are differentiating, and spent cells that will soon be lost. We study the demographics and dynamics of these populations––their sizes, compositions, and spatial distributions over time. Fields of interest: Stem cell biology, epithelial cell and tissue biology, cell-to-organ scale mechanisms, collective cellular regulation. Approaches: Drosophila genetics, in vivo live imaging, quantitative image analysis. |
Detlef Obal https://profiles.stanford.edu/detlef-obal | Anesthesiology, Perioperative and Pain Medicine | We are working on the effect of chronic opioid administration on all aspects of the cardiovascular system. We are using inducible pluripotent stem cells to generate patient specific different types of cardiovascular cells (cardiomyocytes, endothelial cells) and embryonic bodies, to understand how chronic opioid administration changes cellular physiology and G-protein coupled receptor signaling. Beside functional assays, we are also using TIRF microscopy (in collaboration with the Kobilka lab) to further understand the behaviour of GPCRs on primary cardiomyocytes and endothelial cells. In contrast to previous studies, in which receptor systems have been overexpressed in different animal models or rodent derived cells, our experiments will be conducted in a natural environment reflecting the normal physiological behaviour of these receptors human cells. |
Michelle Odden https://profiles.stanford.edu/michelle-odden | Epidemiology & Population Health | Dr. Odden conducts epidemiologic research to improve our understanding of the optimal strategies to promote healthy aging. She works closely with several cohort studies, including the Cardiovascular Health Study and Jackson Heart Study, and has ongoing projects that incorporate electronic health record data from the Veterans Health Administration and the Health Improvment Network. She has a special interest in populations who have been underrepresented in research including the very old, frail, and racial/ethnic minorities. Her work has focused on prevention of cardiovascular and kidney outcomes, as well as preservation of physical and cognitive function in older adults. She is also strongly interested in epidemiological and statistical methods to reduce biases in observational studies, and has expertise in causal inference methods. |
Scott Owen https://profiles.stanford.edu/scott-owen | Neurosurgery | The Owen lab studies how neuromodulation can shape the activity of targeted cell types and circuits to guide behavior. Our goal is to understand how specialized physiology, connectivity, and cell-signaling support the diverse functions of individual neuronal cell types. How can a deeper understanding of these features guide future therapeutic interventions for neurological diseases including Autism, Obsessive-Compulsive Disorder, and Parkinson’s Disease? To address these questions, we integrate a diverse set of experimental approaches, including human and mouse brain slice physiology, single-cell RNA sequencing, operant mouse behavior, in vivo physiology, imaging, and tool development. |
Julia Palacios https://profiles.stanford.edu/julia-palacios | Statistics | Machine learning, Bayesian statistics, statistical genetics, computational biology, quantitative genetics, Biomedical Data Science, Phylodynamics of Infections Diseases. |
Oxana Palesh http://med.stanford.edu/profiles/Oxana_Palesh/ | Psychiatry | Dr. Palesh's Cancer Survivorship Research Lab at Stanford University investigates the impact of cancer treatments on various functions of overall wellbeing. Our research looks at sleep, fatigue, cognition, and neuronal changes associated with quality of life. We focus on understanding the etiology and psychophysiology of treatment side effects in cancer patients and survivors with the goal of developing and testing novel therapeutic approaches to improve clinical outcomes and reduce symptoms, premature aging, and mortality. Our ongoing clinical research includes testing novel behavioral interventions as well as innovative delivery approaches for management of sleep, cancer related fatigue, circadian rhythm disruption, cancer-related cognitive impairments, and health-related quality of life functioning during and subsequent to cancer treatment. We are interested in developing interventions that can also be delivered widely in community oncology settings across the United States, and we are therefore testing these interventions’ efficacy in such settings. |
Victoria N Parikh https://profiles.stanford.edu/victoria-parikh | Assistant Professor of Medicine, Cardiovascular Medicine | I study inherited cardiovascular diseases, mainly focused on cardiomyopathy and arrhythmias. These are the kinds of diseases that cause young people to die suddenly or need heart transplants, so it's incredibly important that we understand not only how to diagnose them accurately but also prevent these feared outcomes. My lab uses a variety of data types and techniques to improve genetic diagnosis and understand mechanisms of these diseases. We integrate clinical disease cohort data with population level genomics and high throughput single cell genome engineering and phenotyping to clarify the disease causality of genetic variants in known genes. We are also particularly interested in how disrupted RNA splicing can cause disease. We use deep phenotyping and CRISPR-based genome engineering of human induced pluripotent stem cell derived cardiomyocytes in tandem with long read (Oxford Nanopore) sequencing to understand the effects of disruptions in splicing homeostasis in the cardiomyocyte. |
Karen Parker https://profiles.stanford.edu/karen-parker | Dept. Psychiatry & Behavioral Sciences | The principal goal of the Parker Lab Social Neurosciences Research Program is to better understand the biology of typical and atypical social functioning across a range of species, and to translate these fundamental insights to drive development of novel diagnostic tools to detect, and precision therapeutics to treat, social deficits in patient populations. |
Anca Mirabela Pasca https://profiles.stanford.edu/anca-pasca | Assistant Professor of Pediatrics/Neonatology | The focus of the lab is to understand the molecular mechanisms underlying neurodevelopmental disorders associated with fetal and neonatal brain injury with the long-term goal of translating the lab’s findings into therapeutics. The research team uses region-specific human brain organoids differentiated from induced pluripotent stem (iPS) cells and employs a multidisciplinary approach involving genetics, molecular and developmental neurobiology to ask questions about brain injury during development. |
Sergiu Pasca https://profiles.stanford.edu/sergiu-pasca | Psychiatry and Behavioral Sciences | A critical challenge in understanding the intricate programs underlying development, assembly and dysfunction of the human brain is the lack of direct access to intact, functioning human brain tissue for detailed investigation by imaging, recording, and stimulation. Our lab is using pluripotent stem cells derived non-invasively from human individuals to generate in a dish specific regions of the human brain in a functional 3D preparation we have developed. We are using months-to-years long brain region-specific organoids or spheroids to understand how neurons find their final position in the brain and how they mature functionally. To investigate how different brain regions talk to each-other in normal and diseased states, we introduced a new approach for in vitro assembly of neural circuits named assembloids. We employ state-of-the-art stem cell biology, genome engineering, imaging and neuroscience approaches to identify the dynamical processes that go awry in neural cells derived from patients with neuropsychiatric disorders, such as autism or schizophrenia, and what should be therapeutically targeted in these conditions. |
Manali Patel https://profiles.stanford.edu/manali-patel?tab=bio | Medicine | Cancer Care Delivery; Health Equity; Value-Based Cancer Care |
Anisha Patel https://profiles.stanford.edu/anisha-patel | Pediatrics | • Community-based participatory research • Randomized controlled trials, policy evaluation, mixed methods research • Prevention of chronic diseases, including childhood obesity • Child nutrition and food insecurity • Schools and early care and education settings • Racial/ethnic and socioeconomic disparities in child health |
Ramasamy Paulmurugan https://profiles.stanford.edu/ramasamy-paulmurugan | Radiology | My research group focuses on developing new imaging assays for studying cellular signal transduction networks in cancer and other diseases. Specifically, we apply our extensive experience in molecular biology to develop several in vivo imaging assays for monitoring basic cellular processes and post-translational modifications of proteins, such as methylation, phosphorylation, sumoylation, and many more. We have developed split-reporter protein complementation systems for various reporter proteins (luciferases, fluorescent proteins, and thymidine kinase) and use them for designing various sensors to study cellular signaling processes. Some of the main applications of these assays include imaging the tumor microenvironment, as well as immune cytokine signaling in cancer and infectious diseases. Other applications where we are currently applying these assays include studying protein-protein interactions involved in estrogen receptor signaling, Nrf2-mediated antioxidant signaling in chemoresistance, p53-sumoylation mediated chemotherapy responses in cancer, NFkB mediated cytokine signaling in cancer, and signaling mechanisms associated with APP and Tau protein sumoylations in Alzheimer’s disease. In cancer therapy, we are establishing microRNA-based reprogramming approaches to sensitizing drug-resistant cancers (breast cancer, hepatocellular carcinoma, and glioma) to commonly used chemotherapies. We mainly target oncogenic and tumor suppressor microRNAs (miR-21, miR-10b, miR-122, and miR-100) that are dysregulated in cancers to improve cancer therapy. To deliver intact miRNAs in vivo, we load miRNAs in PLGA-PEG nanoparticles and use ultrasound-microbubble (US-MB) triggered drug delivery strategies for locoregional enhancement of microRNAs in the tumor bed to improve cancer therapy. We evaluate miRNA delivery strategies in small animal models (mice and rats) and optimize US parameters (cavitation, PRF, mechanical energy, and delivery efficiency) in large animal models (pigs and dogs) to address clinical translational feasibilities. We have shown tremendous progress in this area of research with a number of publications in high impact journals. We recently identified five sense and antisense miRNAs (miR-203, miR-218, antimiR-10b, antimiR-19b, and antimiR-21) through a rigorous analysis of miRNA expression data available in TCGA (GDC) and GEO using a biological basis-driven workflow, where these microRNAs target multiple hallmarks of cancer to improve chemo- and immunotherapies in cancer. In synthetic biology, we recently invented the application of high-pressure microfluidic system in the reconstruction of biomolecules derived from cells (proteins and lipids) along with synthetic sources (phospholipids, polymers, and surfactants) to develop self-assembled nano- and micro-structures that mimic biological membranes for drug delivery applications. As part of this process, we developed biomimetic microbubbles (biMBs) and nanobubbles (biNBs) using tumor cell derived exosomes (TDEs) for cancer immunotherapy applications. In this grant, we apply this novel technology to exploit the natural accumulation of biMBs in the immune organs (LNs, lungs, and spleen) while TDE -targeted biNBs accumulation in the tumor to achieve cancer immunotherapy. In addition, we also work on developing novel intranasal vaccine for Covid-19 and other respiratory diseases. As part of this project, we have developed gold nanostar-coated DNA vaccine for SARS-CoV2 disease by activating mucosal immunity. We also extend this platform delivery system for lung cancer gene therapy applications using suicide gene therapy along tumor suppressor microRNAs as a combination treatment. ACHIEVEMENTS: • Developing multiplex-imaging assays to simultaneously measure histone methylations in various lysine marks of histone proteins. • Developing FDA approved polymer nanoparticles to co-deliver therapeutic sense- and antisense- microRNAs for cancer therapy. • Studying estrogen receptor (ER) α and β cross-talk in breast cancer. • Nrf2-Keap1 antioxidant mechanism in drug resistance and chemotherapy in cancers. • Studying the stemness of cancer cells and cancer stem cells in cancer, and targeting Wnt-Beta catenin and NFkB-Nrf2 signaling to improve cancer chemotherapy • SARS-CoV2 intranasal vaccine |
Gary Peltz https://profiles.stanford.edu/gary-peltz | Stanford University Medical School | My laboratory develops and applies state of the art genetic, genomic and stem cell technologies to its research programs. These methodologies are used to discover the mechanisms mediating disease susceptibility and drug response, and to develop new therapies. As one example, we developed a novel computational genetic analysis method, which has identified over 25 genetic factors affecting susceptibility to drug addiction, chronic pain, infectious diseases, and others . An ongoing effort is now analyzing 10,000 biomedical responses in panels of inbred mouse strains. Single-cell RNA sequencing and metabolic analysis are used to identify developmental and disease-causing pathways. Stem cell-based methods for liver engineering are also used. As examples, the Peltz lab has produced mice with humanized livers that are used to improve drug safety; developed methods to produce human liver organoids from iPSC, which are used for studying the pathogenesis of human genetic liver diseases. |
Claudia K. Petritsch https://med.stanford.edu/profiles/claudia-petritsch | Neurosurgery | >Identifying the underlying causes for cellular plasticity and stemness in brain cancer through analyzing human patient-derived cell cultures and xenografts and genetically engineered mouse models. >Unraveling the mechanisms for theory resistance by defining the cellular architecture and intratumoral heterogeneity of human brain cancer using primary surgical tissue and genetically engineered mouse models. >Engaging with the clinical neurosurgery team to sustain a patient-derived models program for pediatric and adult brain tumors and thereby increase the availability of research tools for the community |
Trung Pham https://profiles.stanford.edu/trung-pham | Pediatrics - Infectious Diseases | We study immunology of infectious diseases and host-microbe interactions. Our research program employs murine infection models and brings together immunology, tissue biology, microbiology, and genetics to uncover fundamental mechanisms of tissue immunity and immunophysiology during persistent bacterial infection. Our goals are to understand: 1) the innate and adaptive immune cellular mechanisms that contain pathogens during persistent infection; 2) how tissue physiological functions, such as tissue repair and nutrient regulation, are maintained during persistent infection; 3) how intracellular bacteria survive innate and adaptive antimicrobial mechanisms in infected tissues. We seek to recruit postdoctoral fellows who are passionate about advancing mechanistic understanding of infection biology and to provide a supportive, diverse environment for fellows to advance their scientific and career development. |
Sharon Pitteri https://profiles.stanford.edu/sharon-pitteri | Radiology | The Pitteri laboratory is focused on the discovery and validation of proteins that can be used as molecular indicators of risk, diagnosis, progression, and recurrence of cancer. Proteomic technologies, predominantly mass spectrometry, are used to identify proteins in the blood that are differentially regulated and/or post-translationally modified with disease state. We are particularly interested in developing and applying workflows to study protein glycosylation which is altered in many disease states. We also investigate additional classes of molecules with diagnostic potential. WUsing human plasma samples, tumor tissue, cancer cell lines, and genetically engineered mouse models, the origins of these proteins are being investigated. A major goal of this research is to define novel molecular signatures for prostate and breast cancers, including particular sub-types of these diseases. This laboratory is also focused on the identification of proteins with expression restricted to the surface of cancer cells which can be used as novel targets for molecular imaging technologies. |
Kathleen Poston https://profiles.stanford.edu/kathleen-poston | Department of Neurology, School of Medicine | Clinical and translational research in cognitive impairment and dementia due to Lewy Body diseases (DLB and PD), with an emphasis on imaging and biomarkers. |
Guillem Pratx https://profiles.stanford.edu/guillem-pratx | Radiation Oncology - Medical Physics | The Physical Oncology Lab is interested in making a lasting impact on translational cancer research by building novel physical tools and methods. Areas of active research include: development of novel technologies for enhancing radiation therapy, and novel molecular imaging techniques for visualizing biological processes non-invasively in vivo. |
Judith Prochaska https://profiles.stanford.edu/judith-prochaska | Medicine / Stanford Prevention Research Center | Dr. Prochaska's research program is leveraging technology (social media, telemedicine) to bring treatment of tobacco use into novel settings and to populations with high prevalence of use and with attention to medical education and clinical practice. |
Jochen Profit https://profiles.stanford.edu/jochen-profit | Pediatrics - Neonatal and Developmental Medicine | The Profit lab focuses on optimizing quality of health care delivery to the most vulnerable patients: infants in the neonatal intensive care unit (NICU) with a strong focus on health equity and care culture. Our research is community participatory and transdisciplinary, including patient and family advocates, clinicians, psychologists, sociologists, economists, health services researchers, biostatisticians, computer scientists, and others. We employ a variety of methods, ranging from randomized clinical trials to qualitative analyses. One of our great strengths is our ability to query linked multi-level data from the California Perinatal Quality Care Collaborative (CPQCC), a multi-stakeholder population based network of ~140 NICUs. Our work in improving quality of care for high-risk infants and their families leads the field and has been recognized nationally and internationally. |
Stanley Qi Associate Professor | Bioengineering | Stanley Qi lab is interested in developing new technologies related to precise genome editing and epigenome engineering. His lab has a strong focus on tool development in neurobiology and gene therapy. His lab created the first nuclease-deactivated Cas9 (dCas9) for targeted gene regulation in cells, as well as a series of CRISPR technologies, including CRISPRi and CRISPRa for targeted gene regulation, epigenome editing, LiveFISH for real-time cell imaging, CRISPR-GO for 3D genome manipulation, CasMINI as a compact CRISPR system for gene therapy, hyperCas12a for multi-gene engineering, and CRISPR antivirals aimed at treating broad RNA viruses. |
Jennifer Raymond https://med.stanford.edu/profiles/jennifer-raymond | Professor of Neurobiology | Our lab investigates the neural mechanisms of learning and memory by integrating in vivo and in vitro physiology with behavioral analysis, techniques such as optogenetics for manipulating neural function, and computational approaches. |
David Rehkopf https://profiles.stanford.edu/david-rehkopf | Epidemiology and Population Health | social determinants of health, real world data, social policies and health, aging |
David Relman https://profiles.stanford.edu/david-relman | Medicine | Human microbiome: assembly in early life, stability, resilience. Human clinical research, microbial ecology, ecological statistics, multiple kinds of molecular measurements, multiple body sites. Applications areas under study include, pregnancy, post-partum maternal microbiome, neonatal microbiome assembly, pediatric IBD, antibiotic perturbation and antibiotic resistance gene ecology, iron and arsenic impact on microbiome |
anthony Ricci anthony-ricci | Otolaryngology | The Riccilab studies cellular based mechanism of peripheral hearing and develops strategies to combat hearing loss associated with aging, noise, drug induced and genetically predisposed. We use electrophysiological and optical approaches to study conversation of mechanical sound waves into electrical signals and also to drive synaptic transmission in the auditory periphery. We use high speed imaging to investigate how sensory hair bundles move in response to mechanical stimulation and how this motion activates ion channels. We investigate both pre and postsynaptic response properties in order to understand the signals being sent to the brain. We also are developing non ototoxic aminoglycosides performing structure activity investigations to monitor drug entry into hair cells as well as drug interactions with mitochondrial ribosomes. And finally, we have developed the ability to use in vivo cochlear imaging to monitor hair cell activity during sound stimulation. |
Thomas Robinson https://profiles.stanford.edu/thomas-robinson | Pediatrics | Impacts of digital technology on health and behavior; Obesity prevention and treatment in children, adolescents and families; Environmental sustainability behaviors; Designing and evaluating, in randomized controlled trials, theory-based individual-, family- and community-based behavioral, social, environmental and policy interventions to improve health and well-being and reduce racial/ethnic and socio-economic inequalities |
Carolyn Ines Rodriguez https://profiles.stanford.edu/carolyn-rodriguez?tab=bio | Psychiatry | Dr. Rodriguez leads studies investigating the brain basis of severe mental disorders. Her landmark clinical trials pioneer rapid-acting treatments for illnesses including Obsessive-Compulsive Disorder (OCD) and Posttraumatic Stress Disorder (PTSD). Her NIH-, foundation-, and donor-funded mechanistic and clinical efficacy studies span targeted glutamatergic and opioid pathway pharmacotherapy, non-invasive brain stimulation, and psychotherapy for OCD, PTSD, and hoarding disorder. |
Patricia Rodriguez Espinosa https://profiles.stanford.edu/patricia-rodriguez-espinosa | Epidemiology and Population Health | The ultimate goal of Dr. Rodriguez Espinosa's research is to decrease health inequities among racial/ethnic minority populations, particularly Latinxs and immigrant communities, through transdisciplinary and community-engaged scholarship. Her research aims to understand factors that create and maintain health inequities (e.g., racial residential segregation) and use these insights to develop novel multi-level interventions and health promotion programs to address the inequity gap and that include multi-sectoral collaborations. Dr. Rodriguez Espinosa's research has also centered around developing the science of Community-Based Participatory Research, citizen science, and other participatory research approaches. |
Kacper Rogala https://profiles.stanford.edu/kacper-rogala | Structural Biology, and Chemical & Systems Biology | Structural and chemical biology of nutrient sensing and nutrient trafficking in cells. We are a team of basic scientists, revealing mechanisms of proteins, and applying that knowledge in the development of novel therapeutics against cancer. |
Rajat Rohatgi https://profiles.stanford.edu/rajat-rohatgi | Department of Biochemistry | Our overall goal is to uncover new regulatory mechanisms in signaling systems, to understand how these mechanisms are damaged in disease states and to devise strategies to repair their function. |
Sherri Rose https://profiles.stanford.edu/sherrirose | Med-PCOR | My research is centered on developing and integrating innovative statistical machine learning approaches to improve human health. Within health policy, I work on risk adjustment, ethical algorithms in health care, comparative effectiveness research, and health program evaluation. |
Elsie Ross https://profiles.stanford.edu/intranet/elsie-ross | Assistant professor of surgery | Improving care for patients with cardiovascular disease by leveraging big data and machine learning to build tools that automate diagnosis and predict outcomes. |
Daniel Rubin https://profiles.stanford.edu/daniel-rubin | Biomedical Data Science, Radiology, and Medicine | The QIAI lab focuses on cutting‐edge research at the intersection of biomedical data science and medicine, developing and applying artificial intelligence (AI) methods to leverage large amounts of medical data for biomedical discovery, precision medicine, and precision health (early detection and prediction of future disease). We develop novel methods for in AI in medicine, particularly for analyzing text and images, including multi-modal and multi-task learning, weak supervision, knowledge representation, NLP, image processing, and decision theory to tackle the challenges of leveraging medical Big Data. Our exciting work is bridging a spectrum of biomedical domains with multidisciplinary collaborations with top scientists at Stanford as well as with other institutions internationally. The QIAI lab provides a unique multidisciplinary environment for conducing innovative AI-based healthcare research with a strong record of scholarly publication and achievement. Core research topics in the laboratory include: (1) automated image annotation using unsupervised methods of processing associated radiology reports using word embeddings and related methods; (2) developing methods of analyzing longitudinal EMR data to predict clinical outcomes and best treatments, e.g., for COVID-19, (3) creating multi-modal deep learning models integrating multi-dimensional medical records data and other data to discover electronic phenotypes of disease, (4) developing AI models with noisy or sparse labels (weak supervision), and cross-modal, multi-task learning, and even novel observational AI approaches by watching users work with data, and (5) developing and implementing algorithms for federated learning of deep learning models that leverage multi-institutional data while avoiding the barriers to data sharing. |
Mirabela Rusu https://profiles.stanford.edu/mirabela-rusu | Radiology | AI, Prostate cancer, Breast Cancer, Radiology-Pathology Integration |
Manish Saggar http://profiles.stanford.edu/manish-saggar | Psychiatry | Network Science, Dynamical Systems, Computational Neuropsychiatry, rsfMRI, TDA, fNIRS |
Gregory Sahlem https://profiles.stanford.edu/gregory-sahlem | Psychiatry | Dr.Sahlem is an Assistant Professor in the Department of Psychiatry and Behavioral Sciences. He is board-certified in general psychiatry and addictions medicine, as well as fellowship-trained in the research and clinical application of neuromodulation-based treatments including repetitive Transcranial Magnetic Stimulation (rTMS), electroconvulsive therapy (ECT), and vagus nerve stimulation (VNS). He additionally has advanced training in the treatment of mood and sleep disorders. In addition to being an active clinician, Dr.Sahlem is a member of the Stanford Brain Stimulation Lab and directs the Addictions Research Section of the Lab. Major areas of study for Dr.Sahlem include: The development of rTMS as a focused treatment for addictive disorders; the development of a novel form of ECT theorized to have reduced cognitive side effects, Focal Electrically Administered Seizure Therapy (FEAST), and; the further development of rTMS for the treatment of mood disorders. While working with Dr.Sahlem you will receive training in the development and execution of clinical trials. You will primarily use neuromodulation based techniques including rTMS, but Dr.Sahlem also has expertise in the research use of pharmacologic and behavioral approaches (and so can provide assistance in the conduct of trials using these forms of intervention). In terms of measurement techniques, you will gain experience in behavioral assessments as well as the use of electroencephalography (EEG) and functional magnetic resonance imaging (fMRI). |
Kathleen Sakamoto https://profiles.stanford.edu/kathleen-sakamoto | Pediatrics | Aberrant hematopoiesis, targeted therapies, signaling, gene regulation, leukemia, bone marrow failure |
Julia Salzman https://profiles.stanford.edu/julia-salzman | Department of Biomedical Data Science | Our group develops new statistical methods for biological inference on fundamental questions in the evolution of genomes, including: What is the mechanism and function for single cell regulation of RNA splicing? What drives genomic innovation and diversity from bacterial to eukaryotes? To approach these questions, we use probabilistic, statistical, and computational biological approaches and test these predictions in experimental systems. Our recent papers highlight some of our work in massive single cell sequencing data. We are committed to fostering an interdisciplinary, inclusive environment and are actively recruiting undergraduates, graduate students, and postdoctoral fellows. |
Peter Santa Maria https://profiles.stanford.edu/peter-luke-santa-maria | Assistant Professor | Infectious disease of the ear, medical devices, drug discovery and therapeutics |
Kavita Sarin https://profiles.stanford.edu/kavita-sarin | Dermatology | Our lab uses next generation sequencing technologies to investigate genetic and molecular alterations associated with skin cancer and skin diseases. Using genetic and molecular data obtained from patients, we predict and develop new therapeutics for a variety of skin disorders. Our ultimate goal is to utilize molecular information to manage and treat patients with dermatologic disorders |
Kavita Sarin https://profiles.stanford.edu/kavita-sarin | Dermatology | Our lab uses next generation sequencing technologies to investigate genetic and molecular alterations associated with skin cancer and skin diseases. Using genetic and molecular data obtained from patients, we predict and develop new therapeutics for a variety of skin disorders. Upcoming projects include a large genetic study to identify genetic modifiers of organ disease manifestations in a genetic disease called Neurofibromatosis Type 1 |
Nazish Sayed https://profiles.stanford.edu/nazish-sayed | Department of Surgery and Cardiovascular Institute | The Sayed Laboratory is focused on the development of novel technologies that drive innovation in regenerative medicine, disease modeling, and drug testing in vascular biology. The lab conducts translational research in vascular biology and aims to understand the role of the vasculature in the development of cardiac diseases, including those due to inherited genetic variants or environmental insults such as type 2 diabetes or hypertension. The lab employs the human induced pluripotent stem cell (iPSC) technology to generate patient-specific vascular cells (endothelial and vascular smooth muscle cells) as an alternative to animal models providing a human tissue surrogate for research that is scalable and sustainable. By employing this unique platform, the lab also investigates the role of chemotherapeutic agents (anti-cancer drugs) on the vasculature. Dr. Sayed’s lab has also established an endothelial regeneration program, where they leverage the innate immune system to regenerate endothelial cells from human fibroblasts. Work from the lab has led to seminal discoveries in the areas of 1) Nitric oxide (NO) biology, (2) vascular biology, (3) stem cell biology, (4) cardiovascular disease modeling (5) cardio-oncology. |
Birgitt Schuele https://profiles.stanford.edu/birgitt-schuele | Pathology, Stanford Medicine | The Schuele lab works on gene discovery and novel stem cell technologies to generate stem cell models from patients and families with Parkinson’s disease to understand the underlying causes of neurodegeneration and to develop biomarkers and new therapies for Parkinson’s disease. We apply iPSC modeling and cell transplantation in vivo, multiplex imaging, cellular phenotyping, and novel genomic technologies. |
Nigam Shah https://profiles.stanford.edu/nigam-shah | Medicine | Data science, AI in Healthcare, Electronic Health Records, Medical Informatics, Predictive Modeling |
Nirao Shah https://profiles.stanford.edu/nirao-shah | Psychiatry | Molecular and neural network mechanisms that control social behaviors in the two sexes in health and disease. Molecular, genetic, circuit, and systems neuroscience. In vivo calcium imaging; in vivo optogenetics; in vivo imaging of hormonally regulated synaptic plasticity; behavioral analysis, genetic engineering of mice, flies, voles. |
Gavin Sherlock https://profiles.stanford.edu/gavin-sherlock | Genetics | Adaptive evolution, genetics networks, antifungal drug resistance, synbio, high throughput sequencing |
Gen Shinozaki https://profiles.stanford.edu/gen-shinozaki | School of Medicine, Department of Psychiatry and Behavioral Sciences | Shinozaki Lab is investigating epigenetic mechanism of neuroinflammation, delirium and aging using human samples as well as mouse model. We are also developing a novel EEG device approach to detect delirium and predict patient outcomes to improve real world clinical practice. With our clinical (real human data), translational (novel device and biomarker development) and basic (epigenetics and animal model) investigation, our goal is to have a better patient care for aging population. |
Julia F Simard https://profiles.stanford.edu/julia-simard | Department of Epidemiology & Population Health | Keywords: Autoimmune disease (primarily rheumatic disease such as systemic lupus), reproductive epidemiology (pregnancy, preeclampsia, preterm delivery), disparities, and misdiagnosis and misclassification. My lab focuses on three main areas: 1. Reproductive epidemiology, largely with a focus on high-risk pregnancy in clinically-complex populations 2. Autoimmune disease epidemiology and outcomes research, largely focused on systemic lupus and related rheumatic autoimmune diagnoses and outcomes ranging from cancer and infection, to mortality. 3. Evaluating implicit bias, misdiagnosis, and clinical decision-making. We use a wide-range of tools and resources from large population-based registers of entire countries or regions (Sweden, Israel, BC Canada) to small clinical cohorts to survey data and randomized experiments. We are interested in the interaction of many of the above areas and extend into evaluating disparities by sex, gender, race, and ethnicity. |
Julia Simard https://profiles.stanford.edu/julia-simard | Epidemiology and Population Health | Autoimmune disease, reproductive and perinatal epidemiology, epidemiology methods, disparities, and bias in medicine |
Laura Simons https://profiles.stanford.edu/laura-simons | Department of Anesthesiology, SOM | The primary goal of this laboratory is to promote the health and well being of children and adolescents with chronic pain and their families. In line with this goal, research projects focus on biological, neurological, cognitive, affective, and social risk and resiliency factors of the pain experience. Projects include brain imaging, longitudinal clinical cohort, and treatment interventions studies. |
Georgios Skiniotis https://profiles.stanford.edu/georgios-skiniotis | Molecular and Cellular Physiology AND Structural Biology | Mechanisms of transmembrane signaling through cell surface receptors with a focus on GPCRs. We use cryoEM, biochemistry, biophysics and computational tools. |
Melody Smith https://profiles.stanford.edu/melody-smith | Division of Blood and Marrow Transplantation and Cellular Therapy | Our lab focuses on the biology of chimeric antigen receptor (CAR) T cells in order to improve the efficacy and safety of this therapy (1) by investigating donor and third-party CAR T cells in an immunocompetent mouse model of allogeneic hematopoietic cell transplant (allo-HCT) and (2) by assessing the impact of the intestinal microbiome on CAR T cell response. We will seek to enhance the development, administration, and mechanistic understanding of how to safely administer donor and third-party CAR T cells with the aim to potentially translate our work to the clinic. We will investigate the regulatory mechanism of the impact of bacterial taxa and the metabolites that they produce on CAR T cell outcomes. |
Mike Snyder https://med.stanford.edu/snyderlab.html | Genetics | Deep data anlaysis of human health, aging cancer. Single cell maps of normal and cancer tissues. Wearables. |
Justin Sonnenburg https://profiles.stanford.edu/justin-sonnenburg | Microbiology and Immunology | Gut microbiota dynamics, function, interaction with the host. Metagenomic sequencing, metabolomics, anaerobic microbiology, gnotobiotic mouse models, immune profiling |
Daniel Spielman https://profiles.stanford.edu/daniel-spielman | Radiology | Medical Imaging, MRI, PET, metabolic imaging |
Kristan Staudenmayer https://profiles.stanford.edu/kristan-staudenmayer | School of Medicine, Department of Surgery | Healthcare systems, healthcare economics, trauma, the elderly, COVID |
Tanya Stoyanova https://profiles.stanford.edu/tanya-stoyanova | Radiology | Stoyanova lab develops new early cancer detection methods and therapeutic strategies for late stage cancers. The current research focus is on protein-based biomarkers for early cancer detection as well as development of new small molecule inhibitors and antibody-based therapies for prostate and other epithelial cancers. The ultimate goals of the laboratory are to improve the early diagnosis and prognosis of clinically significant cancers and guide the development of novel and effective therapeutic strategies for metastatic prostate and other epithelial cancers. |
Aaron Straight https://profiles.stanford.edu/aaron-straight | Biochemistry | Fields of Research: Chromosome Biology, Chromatin, Noncoding RNA, Genome Organization. Technologies: Microscopy, Genomics, Cell Biology, Genome Engineering, Biochemistry |
Brian Suffoletto https://profiles.stanford.edu/brian-suffoletto | Emergency Medicine | My programmatic research is in prevention of alcohol, drug use, and mental health crises in young adults. I am interested in identifying at-risk individuals in health settings (e.g. emergency departments), using digital technology (e.g. smartphones, sensors) to sense periods of risk during everyday life, and applying novel techniques (e.g. micro-interventions) and behavioral strategies (e.g. implementation intentions) to reduce risk of associated harms. |
Katrin Svensson https://profiles.stanford.edu/katrin-svensson | Pathology | The Svensson Laboratory is dedicated to the discovery of new pathways of energy regulation. We are using a combination of secretome proteomics, gene editing and physiology approaches to better understand how to target complex diseases such as aging, metabolic diseases, and cancer. |
Will Talbot https://profiles.stanford.edu/william-talbot | Developmental Biology | We use genetic, molecular, and cellular approaches to investigate the development of the vertebrate nervous system |
Longzhi Tan https://profiles.stanford.edu/longzhi-tan | Neurobiology | The Tan Lab studies the single-cell 3D genome architectural basis of neurodevelopment and aging by developing the next generation of in vivo multi-omic assays and algorithms, and applying them to the human and mouse cerebellum. |
Avnesh S Thakor https://profiles.stanford.edu/avnesh-thakor | Radiology | Fields of Interest: Precision Delivery, Stem cell therapies and microenvironment optimization Technologies: Bioscaffolds, Nanoparticles and focused ultrasound Theme: Translational Regenerative Medicine |
Jennifer A Tremmel https://profiles.stanford.edu/jennifer-tremmel | Associate Professor of Medicine (Cardiovascular) | As director of the Women's Heart Health Program, we conduct a variety of clinical studies aimed at women's cardiovascular health and sex differences in CVD. For example, we are currently doing research in angina and normal coronary arteries (microvascular/endothelial dysfunction and myocardial bridging), spontaneous coronary artery dissection (SCAD), myocardial infarction and normal coronary arteries (MINOCA), pregnancy and heart disease, and psychosocial stress and CVD. |
Tulio Valdez https://profiles.stanford.edu/tulio-valdez?tab=bio | OTOLARYNGOLOGY | IMAGING ,INFECTIOUS DISEASES, |
Capucine Van Rechem https://profiles.stanford.edu/capucine-van-rechem | Pathology | Dr. Van Rechem lab is interested in the molecular impact of chromatin modifiers on disease development, with an emphasis on cancer. Her laboratory undertakes a cell-cycle specific angle to explore specific functions such as gene expression and replication timing. They also explore unconventional direct roles for these factors in the cytoplasm, with a focus on protein synthesis. Their ultimate goal is to provide needed insights into new targeted therapies. |
Alexander Vezeridis https://profiles.stanford.edu/alexander-vezeridis | Radiology | Our lab is the Stanford Interventional Radiology Translational Theranostics (SIRTT) Laboratory, dedicated to developing next generation therapies using minimally invasive image-guided approaches. We are working on making interventional radiology tools and techniques smarter by incorporating advances from computer science, electronics, and materials science. We have an interest in contrast agent developing using techniques in chemistry and nanotechnology. We also operate on the clinical end of the translational spectrum by analyzing how interventional physicians learn and improve. We are very interested in identifying orphan disease conditions not served by other specialties or best treated by locally applied therapies using interventional techniques. We are committed to diversity in the biomedical sciences and welcome your interest in the group. |
Todd Wagner https://profiles.stanford.edu/todd-wagner | Surgery | I am an Associate Professor in the Department of Surgery at Stanford University and Director of the Health Economics Resource Center at the Palo Alto VA. I have worked for 22 years in health care delivery systems where economics and clinical care intersect, raising questions about cost and quality. My research has focused on ways to improve the functioning and efficiency of health care systems. I have mentored over 35 early career investigators |
Kevin Wang https://profiles.stanford.edu/kevin-wang | Dermatology | We are primarily interested in two fundamental areas in biomedical science--how various dynamic epigenetic changes in chromatin structure impact gene expression during stem cell pluripotency/self-renewal, cellular differentiation, and reprogramming, and how the three-dimensional chromosomal structure and its dysregulation contribute to development of diseases such as aging and cancer. As a physician scientist who see complex medical patients in the clinic, the long-term goal of my lab is to translate our understanding of these complex mechanisms to studies of human diseases. |
Xinnan Wang https://profiles.stanford.edu/xinnan-wang?tab=bio | Neurosurgery | Mitochondria move and undergo fission and fusion in all eukaryotic cells. The accurate allocation of mitochondria in neurons is particularly critical due to the significance of mitochondria for ATP supply, Ca++ homeostasis and apoptosis and the importance of these functions to the distal extremities of neurons. In addition, defective mitochondria, which can be highly deleterious to a cell because of their output of reactive oxygen species, need to be repaired by fusing with healthy mitochondria or cleared from the cell. Thus mitochondrial cell biology poses critical questions for all cells, but especially for neurons: how the cell sets up an adequate distribution of the organelle; how it sustains mitochondria in the periphery; and how mitochondria are removed after damage. The goal of our research at the Xinnan Wang Lab is to understand the regulatory mechanisms controlling mitochondrial dynamics and function and the mechanisms by which even subtle perturbations of these processes may contribute to neurodegenerative disorders. |
Sui Wang https://profiles.stanford.edu/sui-wang | Ophthamology | My lab focuses on understanding retinal development and diabetic retinopathy. We utilize genetics, genomics, virology, and molecular and cellular biology approaches to study how different types of retinal cells respond to developmental cues and disease insults, such as diabetes. |
Adam Wang https://profiles.stanford.edu/adamwang | Radiology | The Wang Group develops advanced x-ray and CT imaging systems and methods. Our group's interests revolve around the following areas: - Novel systems and methods for x-ray and CT imaging - Applications of artificial intelligence / machine learning / deep learning to medical imaging - Dual energy / spectral imaging, including photon counting detectors - Monte Carlo and Deterministic methods for x-ray imaging and radiation dose - Model-based image reconstruction |
Matthew Wheeler https://profiles.stanford.edu/matthew-wheeler | Medicine/CV Medicine | My group seeks to uncover the etiology and mechanism of very rare and undiagnosed diseases; genetic cardiomyopathies and genetic striated myopathies; and to understand the molecular determinants of the beneficial effects of exercise. In addition we have an active clinical trials team and a software developer/bioinformatics team with diverse expertise. We collaborate closely with several investigators across Stanford and are active in several consortia. We use genomics, transcriptomics, proteomics, bioinformatics, and human cellular and model organisms to understand mechanisms of variants of uncertain significance and modifiers of Mendelian disease severity. |
John Witte https://profiles.stanford.edu/john-witte | Epidemiology and Population Health | The Witte Lab is a computational and epidemiological research group investigating the genetic and environmental contributions to disease risk and progression. We develop and apply novel genetic epidemiological methods to decipher the mechanisms underlying complex diseases. In particular, we focus on 1) Pleiotropy, assessing the shared genetic basis across cancers and other diseases; 2) Risk Modeling, evaluating disease risk with polygenic risk scores and hierarchical models; and 3) Prostate Cancer: finding genetic risk factors, improving screening, and reducing disparities. |
Sherry Wren https://profiles.stanford.edu/sherry-wren | Surgery | Humanitarian Surgical Care in Conflict Zones - developing a minimal data set for postoperative mortality prediction to inform quality improvement in care. |
Sean M Wu https://profiles.stanford.edu/ming-wu | Cardiovascular Institute - UTL | Heart development, stem cell biology, 3D bioprinting/tissue engineering, single cell RNA sequencing approaches to heart development and cardiac inflammation |
Joseph Wu https://profiles.stanford.edu/joseph-wu | Medicine | stem cell, genomics, disease modeling, genome editing, drug screening, precision medicine, clinical trial in a dish |
Phillip Yang https://profiles.stanford.edu/phillip-yang | Department of Medicine, Professor | Translation of stem cell biology, employing novel molecules, peptides, and organelles discovered from their extra-cellular vesicles, is the focus of our lab. The specific research question is how to repair and regenerate the injured heart tissue. Molecular imaging is employed to facilitate rapid translation of novel discoveries. Advanced molecular and cellular assays are combined with novel imaging tools to validate the findings. |
Fan Yang https://profiles.stanford.edu/fan-yang | Ortho Surg and Bioengineering | The overall goal of our lab's research is to develop novel biomaterials and engineering tools to modulate biology and impact medicine. One focus area of our research is to develop injectable biomaterials to regenerate and rejuvenate human tissues by promoting stem cell differentiation, vascularization or immunomodulation. We are engineering 3D in vitro disease models with in vivo-mimicking phenotype and drug responses. Such 3D models could accelerate discovering novel druggable targets and enable high-throughput drug screening with significantly reduced cost and time. |
Priscilla Yang https://med.stanford.edu/profiles/priscilla-yang | Microbiology and Immunology | My research group focuses on understanding the mechanisms responsible for viral replication and development of new strategies to combat viral pathogens. We combine chemical biology, medicinal chemistry, and molecular virology approaches to tackle challenges in both basic and translational research. Over the past decade, our efforts have centered on two significant problems: first, addressing the challenges that limit our current arsenal of antivirals and second, understanding the specificity and function of host lipids in RNA virus replication. We are keenly interested in discovery of new antiviral targets and strategies and leveraging these discoveries to develop first-in-class small molecule antivirals. We also have a strong interest in developing or adapting tools from chemistry, chemical engineering, and biophysics to probe new areas of virology. |
Jason Yeatman https://profiles.stanford.edu/jason-yeatman | Pediatrics | Our mission is to both use neuroscience as a tool for improving education, and use education as a tool for furthering our understanding of the brain. On the one hand, advances in non-invasive, quantitative brain imaging technologies are opening a new window into the mechanisms that underlie learning. For children with learning disabilities such as dyslexia, we hope to develop personalized intervention programs that are tailored to a child’s unique pattern of brain maturation. On the other hand, interventions provide a powerful tool for understanding how environmental factors shape brain development. Combining neuroimaging with educational interventions we hope to further our understanding of plasticity in the human brain. |
Ellen Yeh yehlab.stanford.edu | Pathology | We study symbioses in understudied protist systems, including the secondary plastid in malaria and a nitrogen-fixing symbionts with diatoms; applications in global and planet health |
Michael Zeineh https://profiles.stanford.edu/michael-zeineh | Radiology | Neuroimaging, neuroradiology, advanced MRI, Alzheimer's disease, mild traumatic brain injury in sports, epilepsy, chronic fatigue syndrome, exosomes |
Michael Zeineh https://profiles.stanford.edu/michael-zeineh | Radiology, Associate Professor | My lab focuses on translating advanced MRI into clinical practice. In Alzheimer’s disease, we are investigating the nature of iron deposition to understand how iron interacts with inflammation, amyloid, and tau in the progression of AD. We bring to this disease the full arsenal of imaging: ultra-high resolution MRI of human AD specimens coupled with novel histological methods including x-ray microscopy and electron microscopy. We bring this armamentarium full circle to living human imaging with 7.0T MR and multi-tracer PET-MR. In mild traumatic brain injury, we are studying the imaging signatures of brain insult that occur in high-contact sports using advanced MRI combined with mouthguard accelerometer measurements of impacts. In chronic fatigue syndrome and long-COVID, we are identifying microstructural changes that accompany fatigue and correlate with systemic circulating cytokines that together may form a biomarker for this disorder. |