Home

We are a diverse group of creative innovators and advocates. We are committed to the development and use of biomedical interventions to address global health concerns.

The Bill & Melinda Gates Medical Research Institute is a non-profit organization dedicated to the development and effective use of novel biomedical interventions addressing substantial global health concerns, for which investment incentives are limited.

The institute works through collaborating partners and organizations, coordinating and driving the full spectrum of biopharmaceutical development activities, including pre-clinical development, full clinical development (from phase 1 through to and including phase 3), and global regulatory interactions. 

The institute focuses on programs aimed at reducing the burden of TB, malaria, diarrheal diseases, and maternal, newborn, and child illnesses worldwide.

As an affiliate of the Bill & Melinda Gates Foundation, the institute’s programs are focused on disease and health areas of primary focus at the foundation.  The interventions under study and development are derived from sources both within and external to the foundation.

Gates MRI Leadership Team

Patrice Matchaba, M.D.

Work with Us

We are a diverse group of passionate change-makers, creative fixers and relentless advocates. We are committed to saving lives by improving the pace and predictability of translational medicine in global health.

Image of gloved hands pipeting

Campus Construction Update

As part of our work to ensure the look of our campus matches the exceptional care you've come to expect, we're closing the corridor between the Moakley and Menino lobbies for approximately one month, starting on Saturday, Aug. 10. Thank you for your patience during this time.

Click here to learn more about our campus redesign. 

Utility Menu

  • Request an Appointment

Popular Searches

  • Adult Primary Care
  • Orthopedic Surgery

boston medical research institute

Clinical Research Network

Research based on equity with a vision to advance medicine for all people

About the CRN

The Clinical Research Network (CRN), one of two Center for Clinical Research Advancement (CCRA) teams, supports BMC's goal of providing innovative, equitable health care and CCRA's goal of advancing it through cutting-edge research that is both responsive to cultural and linguistic differences and inclusive in research participation and study design. To those ends, the CRN plays three roles: promoting community-engaged research values; offering staffing services for institutionally prioritized or under-resourced clinical trials; and nurturing research infrastructure development.

Promote community engagement | Provide CR services | Incubate infrastructure evolution

Our Services

  • Clinical Research Staffing : Supplying BMC’s researchers, sponsors, and the broader BMC community with high-quality staffing services, including recruitment, to activate rapidly and manage our most complex and clinically important studies.  
  • Community Engagement : Engaging with our community members and partners to understand their needs, interests, and beliefs; build trust in research, provide education and resources; and establish hospital infrastructure that supports inclusivity and community-guided research. 

How can we help?

I am a researcher interested in working with the crn.

  • I need help with clinical research staffing
  • I need help with community engagement planning and implementation
  • I need help with rapid activation and study budgeting

I am a community member or patient interested in learning about research at BMC

  • I need help understanding clinical research
  • I would like to find a study that is right for me
  • I would like to know how the CRN is working with my community
  • I would like to partner with the CRN

I am currently participating in a research study and have questions

  • I need to contact my study team with questions
  • I have a question about research participant payments
  • I need to contact the Institutional Review Board (IRB) with questions

Meet the Team  

Research careers with bmc, systems for research sps, sp policy library, education and training, research operations forms, contact the crn.

Want to keep up with the latest news from Boston Medical Center? Sign up for our monthly patient newsletter, Your BMC.

Center for regenerative medicine

boston medical research institute

Discover CReM

Research at CReM

Cores & Protocols

Mentorship & Outreach

CReM in the News!

Financial support.

Human health and development depend on dynamic networks of physical, and functional, interactions between proteins. However, the details of these networks – how they are formed and how they function – are largely unknown.

Upcoming Seminars

Rachel zemans, md, special guests, klaus kaestner, phd, ms.

Thomas and Evelyn Suor Butterworth Professor in Genetics Associate Director, Penn Diabetes Research Center

Smilow Center for Translational Research

University of Pennsylvania Perelman School of Medicine

Philadelphia, PA

Pulin Li, PhD

Eugene Bell Career Development Professor of Tissue Engineering; Core Member, Whitehead Institute

We Are Hiring

Make a donation, research programs.

  • Publications

boston medical research institute

New publication from the Murphy Lab: A specialized population of monocyte-derived tracheal macrophages promote airway epithelial regeneration through a CCR2-dependent mechanism

Click here to read the article.

Alveolar epithelial type I cells (AT1s) line the gas exchange barrier of the distal lung and have been historically challenging to isolate or maintain in cell culture. Here, we engineer a human  in vitro  AT1 model system via directed differentiation of induced pluripotent stem cells (iPSCs). We use primary adult AT1 global transcriptomes to suggest benchmarks and pathways, such as Hippo-LATS-YAP/TAZ signaling, enriched in these cells. Next, we generate iPSC-derived alveolar epithelial type II cells (AT2s) and find that nuclear YAP signaling is sufficient to promote a broad transcriptomic shift from AT2 to AT1 gene programs. The resulting cells express a molecular, morphologic, and functional phenotype reminiscent of human AT1 cells, including the capacity to form a flat epithelial barrier producing characteristic extracellular matrix molecules and secreted ligands. Our results provide an  in vitro  model of human alveolar epithelial differentiation and a potential source of human AT1s.

boston medical research institute

New Publication from the Kotton Lab on the generation of human alveolar epithelial type I cells from pluripotent stem cells

Kotton and colleagues generate human alveolar epithelial type I cells (AT1s) from induced pluripotent stem cells (iPSCs). The resulting cells can be grown as 3D organoids or in 2D air-liquid interface cultures, displaying many of the molecular, morphologic, and functional phenotypes of primary AT1s.

Click here to see the article

boston medical research institute

CReM Researchers Awarded $14 Million to Understand and Treat Genetic Lung Diseases

A team of researchers led by Darrell N. Kotton, MD, the David C. Seldin Professor of Medicine, has been awarded a five-year, $14 million grant from the NIH’s National Heart, Lung, and Blood Institute (NHLBI) for their research, “Developing Pluripotent Stem Cells to Model and Treat Lung Disease.” The new award will fund an integrated, multi-investigator program project grant where four interacting labs headed by four physician-scientists, all located in the Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, will develop next generation stem cell-based therapies for currently incurable genetic lung diseases affecting children and adults, including childhood and adult interstitial lung diseases, an inherited form of emphysema, cystic fibrosis and primary ciliary dyskinesia.

Generation and directed differentiation of patient-specific ABCA3 mutant and syngeneic gene-corrected iPSC lines produces SFTPCtdTomato-expressing iAEC2s.

The Kotton Lab publishes new paper on iPSC modeling of childhood interstitial lung disease caused by ABCA3 mutations

Mutations in ATP-binding cassette A3 (ABCA3), a phospholipid transporter critical for surfactant homeostasis in pulmonary alveolar type II epithelial cells (AEC2s), are the most common genetic causes of childhood interstitial lung disease (chILD). Treatments for patients with pathological variants of ABCA3 mutations are limited, in part due to a lack of understanding of disease pathogenesis resulting from an inability to access primary AEC2s from affected children. Here, we report the generation of AEC2s from affected patient induced pluripotent stem cells (iPSCs) carrying homozygous versions of multiple ABCA3 mutations. We generated syngeneic CRISPR/Cas9 gene-corrected and uncorrected iPSCs and ABCA3-mutant knockin ABCA3:GFP fusion reporter lines for in vitro disease modeling. We observed an expected decreased capacity for surfactant secretion in ABCA3-mutant iPSC-derived AEC2s (iAEC2s), but we also found an unexpected epithelial-intrinsic aberrant phenotype in mutant iAEC2s, presenting as diminished progenitor potential, increased NFκB signaling, and the production of pro-inflammatory cytokines. The ABCA3:GFP fusion reporter permitted mutant-specific, quantifiable characterization of lamellar body size and ABCA3 protein trafficking, functional features that are perturbed depending on ABCA3 mutation type. Our disease model provides a platform for understanding ABCA3 mutation–mediated mechanisms of alveolar epithelial cell dysfunction that may trigger chILD pathogenesis.

Click here to read the article

boston medical research institute

Latest publication from the Murphy Lab featured on the cover of Blood Advances: De Novo Hematopoiesis from the Fetal Lung!

Hemogenic endothelial cells (HECs) are specialized cells that undergo endothelial-to-hematopoietic transition (EHT) to give rise to the earliest precursors of hematopoietic progenitors that will eventually sustain hematopoiesis throughout the lifetime of an organism. Although HECs are thought to be primarily limited to the aorta-gonad-mesonephros (AGM) during early development, EHT has been described in various other hematopoietic organs and embryonic vessels. Though not defined as a hematopoietic organ, the lung houses many resident hematopoietic cells, aids in platelet biogenesis, and is a reservoir for hematopoietic stem and progenitor cells (HSPCs). However, lung HECs have never been described. Here, we demonstrate that the fetal lung is a potential source of HECs that have the functional capacity to undergo EHT to produce de novo HSPCs and their resultant progeny. Explant cultures of murine and human fetal lungs display adherent endothelial cells transitioning into floating hematopoietic cells, accompanied by the gradual loss of an endothelial signature. Flow cytometric and functional assessment of fetal-lung explants showed the production of multipotent HSPCs that expressed the EHT and pre-HSPC markers EPCR, CD41, CD43, and CD44. scRNA-seq and small molecule modulation demonstrated that fetal lung HECs rely on canonical signaling pathways to undergo EHT, including TGFβ/BMP, Notch, and YAP. Collectively, these data support the possibility that post-AGM development, functional HECs are present in the fetal lung, establishing this location as a potential extramedullary site of de novo hematopoiesis.

boston medical research institute

New publication from the Murphy Lab in Science Advances! They led a Team demonstrating that immune cells drive NET tumor progression and susceptibility to therapies

Neuroendocrine tumors (NETs) are rare cancers that most often arise in the gastrointestinal tract and pancreas. The fundamental mechanisms driving gastroenteropancreatic (GEP)–NET growth remain incompletely elucidated; however, the heterogeneous clinical behavior of GEP-NETs suggests that both cellular lineage dynamics and tumor microenvironment influence tumor pathophysiology. Here, we investigated the single-cell transcriptomes of tumor and immune cells from patients with gastroenteropancreatic NETs. Malignant GEP-NET cells expressed genes and regulons associated with normal, gastrointestinal endocrine cell differentiation, and fate determination stages. Tumor and lymphoid compartments sparsely expressed immunosuppressive targets commonly investigated in clinical trials, such as the programmed cell death protein–1/programmed death ligand–1 axis. However, infiltrating myeloid cell types within both primary and metastatic GEP-NETs were enriched for genes encoding other immune checkpoints, including VSIR (VISTA), HAVCR2 (TIM3), LGALS9 (Gal-9), and SIGLEC10. Our findings highlight the transcriptomic heterogeneity that distinguishes the cellular landscapes of GEP-NET anatomic subtypes and reveal potential avenues for future precision medicine therapeutics.

boston medical research institute

Kotton Lab featured in Boston University Article on Lung Disease Research

For more than 20 years, a team of Boston University scientists have been on a quest to not just figure out how to treat incurable lung diseases, but also how to regenerate damaged lungs so they’re as good as new.

That is the goal of pulmonologist Darrell Kotton and his lab at the Center for Regenerative Medicine (CReM), a joint effort between the University and Boston Medical Center, BU’s primary teaching hospital. By refining their work using sophisticated stem cell technology, Kotton and his team are closer to realizing that vision than ever before.

In two new studies published in Cell Stem Cell, BU researchers detail how they engineered lung stem cells and successfully transplanted them into injured lungs of mice. Two lines of cells targeted two different parts of the lung: the airways, including the trachea and bronchial tubes, and the alveoli, the delicate air sacs that deliver oxygen to the bloodstream. Their findings could eventually lead to new ways for treating lung diseases, including severe cases of COVID-19, emphysema, pulmonary fibrosis, and cystic fibrosis, a disease caused by a genetic mutation.

boston medical research institute

The Mostoslavsky Lab Publishes New Platform to Make T Cells from iPSCs

A robust method of producing mature T cells from iPSCs is needed to realize their therapeutic potential. NOTCH1 is known to be required for the production of hematopoietic progenitor cells with T cell potential in vivo. Here we identify a critical window during mesodermal differentiation when Notch activation robustly improves access to definitive hematopoietic progenitors with T/NK cell lineage potential. Low-density progenitors on either OP9-hDLL4 feeder cells or hDLL4-coated plates favored T cell maturation into TCRab+CD3+CD8+ cells that express expected T cell markers, upregulate activation markers, and proliferate in response to T cell stimulus. Single-cell RNAseq shows Notch activation yields a 6-fold increase in multi-potent hematopoietic progenitors that follow a developmental trajectory toward T cells with clear similarity to post-natal human thymocytes. We conclude that early mesodermal Notch activation during hematopoietic differentiation is a missing stimulus with broad implications for producing hematopoietic progenitors with definitive characteristics.

Click here to access the full article

Centenarian Painting

New Publication for the Murphy Lab, A ‘Blueprint’ for Longevity feature in USA Today, the New York Post and 75 other Media Outlets

Age-related changes in immune cell composition and functionality are associated with multimorbidity and mortality. However, many centenarians delay the onset of aging-related disease suggesting the presence of elite immunity that remains highly functional at extreme old age.

To identify immune-specific patterns of aging and extreme human longevity, we analyzed novel single cell profiles from the peripheral blood mononuclear cells (PBMCs) of a random sample of 7 centenarians (mean age 106) and publicly available single cell RNA-sequencing (scRNA-seq) datasets that included an additional 7 centenarians as well as 52 people at younger ages (20–89 years).

The analysis confirmed known shifts in the ratio of lymphocytes to myeloid cells, and noncytotoxic to cytotoxic cell distributions with aging, but also identified significant shifts from CD4+ T cell to B cell populations in centenarians suggesting a history of exposure to natural and environmental immunogens. We validated several of these findings using flow cytometry analysis of the same samples. Our transcriptional analysis identified cell type signatures specific to exceptional longevity that included genes with age-related changes (e.g., increased expression of STK17A, a gene known to be involved in DNA damage response) as well as genes expressed uniquely in centenarians’ PBMCs (e.g., S100A4, part of the S100 protein family studied in age-related disease and connected to longevity and metabolic regulation).

Collectively, these data suggest that centenarians harbor unique, highly functional immune systems that have successfully adapted to a history of insults allowing for the achievement of exceptional longevity.

Click here to see the article Click here to see the USA Today Article

boston medical research institute

The latest publication from the Kotton lab detailing the transcriptomic programs of iPSC-derived alveolar cells

Dysfunction of alveolar epithelial type 2 cells (AEC2s), the facultative progenitors of lung alveoli, is implicated in pulmonary disease pathogenesis, highlighting the importance of human in vitro models. However, AEC2-like cells in culture have yet to be directly compared to their in vivo counterparts at single-cell resolution. Here, we performed head-to-head comparisons among the transcriptomes of primary (1°) adult human AEC2s, their cultured progeny, and human induced pluripotent stem cell–derived AEC2s (iAEC2s). We found each population occupied a distinct transcriptomic space with cultured AEC2s (1° and iAEC2s) exhibiting similarities to and differences from freshly purified 1° cells. Across each cell type, we found an inverse relationship between proliferative and maturation states, with preculture 1° AEC2s being most quiescent/mature and iAEC2s being most proliferative/least mature. Cultures of either type of human AEC2s did not generate detectable alveolar type 1 cells in these defined conditions; however, a subset of iAEC2s cocultured with fibroblasts acquired a transitional cell state described in mice and humans to arise during fibrosis or following injury. Hence, we provide direct comparisons of the transcriptomic programs of 1° and engineered AEC2s, 2 in vitro models that can be harnessed to study human lung health and disease.

boston medical research institute

The Center for Regenerative Medicine
670 Albany St
2nd Floor
Boston
MA 02118
Boston University & Boston Medical Center

Quick links

Research at the crem, cores & protocols.

© 2021 CReM. All Rights Reserved.

Privacy policy             Terms of use

boston medical research institute

Welcome to CReM

My name is Gabrielle. I'm the Administrative Assistant at CReM. Leave us a short message down below. We will get back to you ASAP!

The Wilson lab is focused on two major aspects of regenerative medicine:

1) Developing gene therapy approaches for the study and treatment of lung diseases: The ability to manipulate gene expression in specified lung cell populations has both experimental and therapeutic potential for lung disease. By developing viral vectors that transduce specific lung cell types in vivo, we hope to minimize potential off-target effects while maximizing our ability to target diseased cell populations. We work with lentiviral and AAV vectors to overexpress or knockdown expression of genes important to disease pathogenesis in the lung.

2) Utilizing induced pluripotent stem cells (iPSC) to study human lung and liver diseases: The Wilson lab is interested in the application of patient-derived iPS cells for the study of lung and liver diseases, such as alpha-1 antitrypsin deficiency (AATD).

The Hawkins Lab is interested in how the human lung develops and responds to injury to better understand human lung disease. Induced pluripotent stem cells (iPSCs) offer a unique opportunity to model human lung disease and bridge the gap between research in animal models and humans.

Using this iPSC platform, we are focused on understanding the molecular mechanisms that control human lung development. We hope to apply this knowledge to advance our understanding of and develop precision medicine approaches for lung disease.

The Murphy laboratory is composed of dynamic and passionate researchers who utilize multiple stem cell-based platforms to answer basic biological questions and combat disease. Central directions of the laboratory include: developmental hematopoiesis, the modeling of blood-borne disease, and discovery and therapeutic intervention in sickle cell disease, amyloidosis, and aging.

The Murphy Lab has pioneered: The world’s largest sickle cell disease-specific iPSC library and platforms and protocols that can used to recapitulate hematopoietic ontogeny and to develop and validate novel therapeutic strategies for the disease; The successful modeling of a protein folding disorder called familial amyloidosis demonstrating the ability to model a long-term, complex, multisystem disease in a relatively short time, using lineage-specified cells (hepatic, cardiac and neuronal) derived from patient-specific stem cells; The first iPSC library created from subjects with exceptional longevity (centenarians) that serves as an unlimited resource of biomaterials to fuel the study of aging and the development of novel therapeutics for aging-related disease.

www.murphylaboratory.com

@DRGJMurphy

The Serrano Lab studies neurodevelopment and cardiovascular development in the context of rare multi-systemic disorders originated by pathogenic variants in epigenetic modifiers like KMT2D.   

We aim to identify shared molecular and cellular mechanisms driving cardiovascular and brain development with particular interest in cell differentiation, migration, and cell cycle progression.   

Our lab combines rare disease modeling in zebrafish together with cardiovascular and neurobiology techniques and human iPSC-derived brain organoids and endothelial cells.   

We believe that a patient-forward focus to our projects will help us to get better understanding of disease mechanisms through basic science research. To this end, we are active in the collaborative community among field experts and rare disease patient-advocacy groups who drive our research program to identify therapeutic targets in patient-specific iPS cells.  

The Mostoslavsky Lab is a basic science laboratory in the Section of Gastroenterology in the Department of Medicine at Boston University.

Our goal is to advance our understanding of stem cell biology with a focus on their genetic manipulation via gene transfer and their potential use for stem cell-based therapy.

The Mostoslavsky’s Lab designed and constructed the STEMCCA vector for the generation of iPS cells, a tool that has become the industry standard for nuclear reprogramming. Project areas in the lab focuses on the use of different stem cell populations, including embryonic stem cells, induced Pluripotent Stem (iPS) cells, hematopoietic stem cells and intestinal stem cells and their genetic manipulation by lentiviral vectors.

Our laboratory have already established a large library of disease-specific iPS cells with a particular interest in utilizing iPS cells to model diseases of the liver, the gastrointestinal tract, prion-mediated neurodegenerative diseases and immune-based inflammatory conditions, using iPSC-derived microglia, macrophages and T/NK cells.

The Gouon-Evans lab investigates cellular and molecular mechanisms driving liver development, regeneration and cancer. We specifically interrogate the role of progenitor/stem cells and how they share similar molecular signature and functions during these 3 processes.

Our innovative tools include: 1) directed differentiation of human pluripotent stem cells (PSC) to generate in vitro liver progenitors and their derivative hepatocytes, the main functional cell type of the liver, 2) mouse models with lineage tracing strategy to track in vivo the fate of progenitor cells, 3) PSC derivative cell transplantation into mouse models with damaged livers as cell therapy for liver diseases, 3) dissection of liver cancer specimens from patients to identify and define the impact of specific cancer stem cells in liver oncogenesis.

Projects in the Gouon-Evans lab will lead to a better understanding of the liver development, to the establishment of multi-modular approaches for improving liver regeneration with PSC derivatives, and will reveal the impact of specific cancer stem cells as a target for diagnosis and therapy in liver oncogenesis.

IMAGES

  1. The NIH Clinical Center

    boston medical research institute

  2. Boston Scientific Medical Research Facility

    boston medical research institute

  3. Toshiko Mori and Maya Lin create buildings for Boston pharmaceutical

    boston medical research institute

  4. Determining Research Institution

    boston medical research institute

  5. Boston Scientific Medical Research Facility

    boston medical research institute

  6. Transactions: HTA adds to its Boston bio portfolio

    boston medical research institute

COMMENTS

  1. Boston Medical Center is a national leader in research

    Guiding knowledge with compassion. At Boston Medical Center, our mission is to deliver the best care possible to better the lives and wellbeing of all our patients. With sophisticated, inclusive, and patient-centered research, we believe we can improve health outcomes and drive change and innovation while prioritizing integrity and trust building.

  2. Homepage - The Bill & Melinda Gates Medical Research Institute

    The Bill & Melinda Gates Medical Research Institute is a non-profit organization dedicated to the development and effective use of novel biomedical interventions addressing substantial global health concerns.

  3. Research Centers and Programs | Boston Medical Center

    Boston Medical Center is committed to empowering all patients to thrive, through our innovative and equitable care. In addition to this commitment, our leadership in health equity, research, and teaching is driving the future of care.

  4. About Us - The Bill & Melinda Gates Medical Research Institute

    The Bill & Melinda Gates Medical Research Institute is a non-profit organization dedicated to the development and effective use of novel biomedical interventions addressing substantial global health concerns, for which investment incentives are limited.

  5. Bill & Melinda Gates Medical Research Institute - Wikipedia

    The Bill & Melinda Gates Medical Research Institute (Gates MRI) is a non-profit biotechnology organization founded with the aim of bringing technologies and strategies to bear on the main health problems of the poor in low-income countries.

  6. Clinical Research Network | Boston Medical Center

    The Clinical Research Network is committed to community-engaged research and provides staffing solutions for institutionally prioritized and under-resourced BMC studies.

  7. Bill & Melinda Gates Medical Research Institute | LinkedIn

    The Bill & Melinda Gates Medical Research Institute is a non-profit organization dedicated to the development and effective use of novel biomedical interventions addressing substantial global...

  8. Research | Harvard Medical School

    This work takes place on the School’s Boston campus and across the metropolitan area at 15 affiliated hospitals and research institutes. Faculty members and students collaborate across the community, pooling resources and expertise to tackle tough problems ranging from understanding the root causes of cancer to developing therapies and ...

  9. Dana–Farber Cancer Institute - Wikipedia

    Dana-Farber Cancer Institute (DFCI) is a comprehensive cancer treatment and research center in Boston, Massachusetts.

  10. CReM - Center for Regenerative Medicine

    The Center for Regenerative Medicine (CReM) is a joint effort between Boston University and Boston Medical Center that brings together nine principal investigators addressing various aspects of developmental biology, stem cells, regeneration and injury, cell lineage specification and disease modeling with a major focus on induced Pluripotent ...