Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Matthew E. Brown is active.

Publication


Featured researches published by Matthew E. Brown.


Blood | 2011

Human lymphoblastoid B cell lines reprogrammed to EBV-free induced pluripotent stem cells

Deepika Rajesh; Sarah J. Dickerson; Junying Yu; Matthew E. Brown; James A. Thomson; Nicholas Seay

Generation of patient-specific induced pluripotent cells (iPSCs) holds great promise for regenerative medicine. Epstein-Barr virus immortalized lymphoblastoid B-cell lines (LCLs) can be generated from a minimal amount of blood and are banked worldwide as cellular reference material for immunologic or genetic analysis of pedigreed study populations. We report the generation of iPSCs from 2 LCLs (LCL-iPSCs) via a feeder-free episomal method using a cocktail of transcription factors and small molecules. LCL-derived iPSCs exhibited normal karyotype, expressed pluripotency markers, lost oriP/EBNA-1 episomal vectors, generated teratomas, retained donor identity, and differentiated in vitro into hematopoietic, cardiac, neural, and hepatocyte-like lineages. Significantly, although the parental LCLs express viral EBNA-1 and other Epstein-Barr virus latency-related elements for their survival, their presence was not detectable in LCL-iPSCs. Thus, reprogramming LCLs could offer an unlimited source for patient-specific iPSCs.


Stem cell reports | 2015

Nonirradiated NOD,B6.SCID Il2rγ−/− KitW41/W41 (NBSGW) Mice Support Multilineage Engraftment of Human Hematopoietic Cells

Brian E. McIntosh; Matthew E. Brown; Bret M. Duffin; John P. Maufort; David T. Vereide; Igor I. Slukvin; James A. Thomson

Summary In this study, we demonstrate a newly derived mouse model that supports engraftment of human hematopoietic stem cells (HSCs) in the absence of irradiation. We cross the NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) strain with the C57BL/6J-KitW-41J/J (C57BL/6.KitW41) strain and engraft, without irradiation, the resulting NBSGW strain with human cord blood CD34+ cells. At 12-weeks postengraftment in NBSGW mice, we observe human cell chimerism in marrow (97% ± 0.4%), peripheral blood (61% ± 2%), and spleen (94% ± 2%) at levels observed with irradiation in NSG mice. We also detected a significant number of glycophorin-A-positive expressing cells in the developing NBSGW marrow. Further, the observed levels of human hematopoietic chimerism mimic those reported for both irradiated NSG and NSG-transgenic strains. This mouse model permits HSC engraftment while avoiding the complicating hematopoietic, gastrointestinal, and neurological side effects associated with irradiation and allows investigators without access to radiation to pursue engraftment studies with human HSCs.


Science Translational Medicine | 2015

Bioengineered vocal fold mucosa for voice restoration

Changying Ling; Qiyao Li; Matthew E. Brown; Yo Kishimoto; Yutaka Toya; Erin E. Devine; Kyeong-Ok Choi; Kohei Nishimoto; Ian G. Norman; Tenzin Tsegyal; Jack J. Jiang; William J. Burlingham; Sundaram Gunasekaran; Lloyd M. Smith; Brian L. Frey; Nathan V. Welham

Primary human vocal fold mucosal cells recapitulate native physiologic function, offering voice restoration to patients with advanced laryngeal disease. Getting vocal about tissue engineering The power of the voice cannot be disputed. For instance, Adele’s lyrics would not elicit chills (or tears) without strategic pitch and harmonizing known as appoggiatura; the chant “Yes we can” garnered more than 69 million popular votes to win Obama the 2008 presidential election; and, more simply, voice is the primary means we all use to communicate with co-workers, loved ones, and the rest of society. Dysphonia—or difficulty speaking from vocal fold tissue damage or loss—can impair one’s ability to be an effective communicator. To provide a new option for those with dysphonia, Ling et al. used two different types of human vocal fold cells to create a functional mucosa. When grafted into the dog larynx ex vivo, the engineered vocal fold reproduced natural physiology, including the vibrations necessary to transmit sound. In vivo, in humanized mice, the engineered mucosa was tolerated by functional human immune cells. These data suggest feasibility for transplant and survival in the larynx as well as for function, ultimately giving patients back their voices. Patients with voice impairment caused by advanced vocal fold (VF) fibrosis or tissue loss have few treatment options. A transplantable, bioengineered VF mucosa would address the individual and societal costs of voice-related communication loss. Such a tissue must be biomechanically capable of aerodynamic-to-acoustic energy transfer and high-frequency vibration and physiologically capable of maintaining a barrier against the airway lumen. We isolated primary human VF fibroblasts and epithelial cells and cocultured them under organotypic conditions. The resulting engineered mucosae showed morphologic features of native tissue, proteome-level evidence of mucosal morphogenesis and emerging extracellular matrix complexity, and rudimentary barrier function in vitro. When grafted into canine larynges ex vivo, the mucosae generated vibratory behavior and acoustic output that were indistinguishable from those of native VF tissue. When grafted into humanized mice in vivo, the mucosae survived and were well tolerated by the human adaptive immune system. This tissue engineering approach has the potential to restore voice function in patients with otherwise untreatable VF mucosal disease.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Functional characterization of human pluripotent stem cell-derived arterial endothelial cells

Jue Zhang; Li-Fang Chu; Zhonggang Hou; Michael P. Schwartz; Timothy A. Hacker; Vernella Vickerman; Scott Swanson; Ning Leng; Bao Kim Nguyen; Angela L. Elwell; Jennifer M. Bolin; Matthew E. Brown; Ron Stewart; William J. Burlingham; William L. Murphy; James A. Thomson

Significance Generating fully functional arterial endothelial cells is a critical problem for vascular development and disease research. Currently, the arterial endothelial cells derived from human pluripotent stem cells lack the range of arterial-specific functions in vitro and the protective function for ischemic tissues in vivo. Here, we combine single-cell RNA sequencing and CRISPR-Cas9 technology to identify pathways for regulating arterial endothelial cell differentiation. We then manipulate these pathways and generate arterial endothelial cells that demonstrate unprecedented arterial-specific functions as well as improve survival of myocardial infarction. These findings facilitate the understanding of vascular development and disease and provide a source of cells that have broad applications for vascular disease modeling and regenerative medicine. Here, we report the derivation of arterial endothelial cells from human pluripotent stem cells that exhibit arterial-specific functions in vitro and in vivo. We combine single-cell RNA sequencing of embryonic mouse endothelial cells with an EFNB2-tdTomato/EPHB4-EGFP dual reporter human embryonic stem cell line to identify factors that regulate arterial endothelial cell specification. The resulting xeno-free protocol produces cells with gene expression profiles, oxygen consumption rates, nitric oxide production levels, shear stress responses, and TNFα-induced leukocyte adhesion rates characteristic of arterial endothelial cells. Arterial endothelial cells were robustly generated from multiple human embryonic and induced pluripotent stem cell lines and have potential applications for both disease modeling and regenerative medicine.


American Journal of Transplantation | 2017

Th17 Responses to Collagen Type V, kα1-Tubulin, and Vimentin Are Present Early in Human Development and Persist Throughout Life

Jeremy A. Sullivan; Ewa Jankowska-Gan; Subramanya Hegde; Matthew A Pestrak; Vrushali V. Agashe; Arick C. Park; Matthew E. Brown; John F. Kernien; David S. Wilkes; Dixon B. Kaufman; Daniel S. Greenspan; William J. Burlingham

T helper 17 (Th17)–dependent autoimmune responses can develop after heart or lung transplantation and are associated with fibro‐obliterative forms of chronic rejection; however, the specific self‐antigens involved are typically different from those associated with autoimmune disease. To investigate the basis of these responses, we investigated whether removal of regulatory T cells or blockade of function reveals a similar autoantigen bias. We found that Th17 cells specific for collagen type V (Col V), kα1‐tubulin, and vimentin were present in healthy adult peripheral blood mononuclear cells, cord blood, and fetal thymus. Using synthetic peptides and recombinant fragments of the Col V triple helical region (α1[V]), we compared Th17 cells from healthy donors with Th17 cells from Col V–reactive heart and lung patients. Although the latter responded well to α1(V) fragments and peptides in an HLA‐DR–restricted fashion, Th17 cells from healthy persons responded in an HLA‐DR–restricted fashion to fragments but not to peptides. Col V, kα1‐tubulin, and vimentin are preferred targets of a highly conserved, hitherto unknown, preexisting Th17 response that is MHC class II restricted. These data suggest that autoimmunity after heart and lung transplantation may result from dysregulation of an intrinsic mechanism controlling airway and vascular homeostasis.


Transplantation Reviews | 2016

Modulation of human allogeneic and syngeneic pluripotent stem cells and immunological implications for transplantation

Sara Dutton Sackett; Matthew E. Brown; Daniel M. Tremmel; Thomas M. Ellis; William J. Burlingham; Jon S. Odorico

Tissues derived from induced pluripotent stem cells (iPSCs) are a promising source of cells for building various regenerative medicine therapies; from simply transplanting cells to reseeding decellularized organs to reconstructing multicellular tissues. Although reprogramming strategies for producing iPSCs have improved, the clinical use of iPSCs is limited by the presence of unique human leukocyte antigen (HLA) genes, the main immunologic barrier to transplantation. In order to overcome the immunological hurdles associated with allogeneic tissues and organs, the generation of patient-histocompatible iPSCs (autologous or HLA-matched cells) provides an attractive platform for personalized medicine. However, concerns have been raised as to the fitness, safety and immunogenicity of iPSC derivatives because of variable differentiation potential of different lines and the identification of genetic and epigenetic aberrations that can occur during the reprogramming process. In addition, significant cost and regulatory barriers may deter commercialization of patient specific therapies in the short-term. Nonetheless, recent studies provide some evidence of immunological benefit for using autologous iPSCs. Yet, more studies are needed to evaluate the immunogenicity of various autologous and allogeneic human iPSC-derived cell types as well as test various methods to abrogate rejection. Here, we present perspectives of using allogeneic vs. autologous iPSCs for transplantation therapies and the advantages and disadvantages of each related to differentiation potential, immunogenicity, genetic stability and tumorigenicity. We also review the current literature on the immunogenicity of syngeneic iPSCs and discuss evidence that questions the feasibility of HLA-matched iPSC banks. Finally, we will discuss emerging methods of abrogating or reducing host immune responses to PSC derivatives.


Chimerism | 2015

No irradiation required: The future of humanized immune system modeling in murine hosts

Brian E. McIntosh; Matthew E. Brown

ABSTRACT Immunocompromised mice are an essential tool for human xenotransplantation studies, including human haematopoietic stem cell (HSC) biology research. Over the past 35 years, there have been many advances in the development of these mouse models, offering researchers increasingly sophisticated options for creating clinically relevant mouse-human chimeras. This addendum article will focus on our recent development of the “NSGW” mouse, which, among other beneficial traits, is genetically modified to obviate the need for myeloablative irradiation of the animals. Thus, the complicating haematopoietic, gastrointestinal, and neurological side effects associated with irradiation are avoided and investigators without access to radiation sources are enabled to pursue engraftment studies with human HSCs. We will also discuss the topics of transgenics, knock-ins, and other mutants with an overarching goal of enhancing chimerism in these animal models.


Stem cell reports | 2018

A Humanized Mouse Model Generated Using Surplus Neonatal Tissue

Matthew E. Brown; Ying Zhou; Brian E. McIntosh; Ian G. Norman; Hannah E. Lou; Mitch Biermann; Jeremy A. Sullivan; Timothy J. Kamp; James A. Thomson; Petros V. Anagnostopoulos; William J. Burlingham

Summary Here, we describe the NeoThy humanized mouse model created using non-fetal human tissue sources, cryopreserved neonatal thymus and umbilical cord blood hematopoietic stem cells (HSCs). Conventional humanized mouse models are made by engrafting human fetal thymus and HSCs into immunocompromised mice. These mice harbor functional human T cells that have matured in the presence of human self-peptides and human leukocyte antigen molecules. Neonatal thymus tissue is more abundant and developmentally mature and allows for creation of up to ∼50-fold more mice per donor compared with fetal tissue models. The NeoThy has equivalent frequencies of engrafted human immune cells compared with fetal tissue humanized mice and exhibits T cell function in assays of ex vivo cell proliferation, interferon γ secretion, and in vivo graft infiltration. The NeoThy model may provide significant advantages for induced pluripotent stem cell immunogenicity studies, while bypassing the requirement for fetal tissue.


Scientific Reports | 2018

Extracellular matrix scaffold and hydrogel derived from decellularized and delipidized human pancreas

Sara Dutton Sackett; Daniel M. Tremmel; Fengfei Ma; Austin K. Feeney; Rachel M. Maguire; Matthew E. Brown; Ying Zhou; Xiang Li; Cori O’Brien; Lingjun Li; William J. Burlingham; Jon S. Odorico

Extracellular matrix (ECM) plays an important developmental role by regulating cell behaviour through structural and biochemical stimulation. Tissue-specific ECM, attained through decellularization, has been proposed in several strategies for tissue and organ replacement. Decellularization of animal pancreata has been reported, but the same methods applied to human pancreas are less effective due to higher lipid content. Moreover, ECM-derived hydrogels can be obtained from many decellularized tissues, but methods have not been reported to obtain human pancreas-derived hydrogel. Using novel decellularization methods with human pancreas we produced an acellular, 3D biological scaffold (hP-ECM) and hydrogel (hP-HG) amenable to tissue culture, transplantation and proteomic applications. The inclusion of a homogenization step in the decellularization protocol significantly improved lipid removal and gelation capability of the resulting ECM, which was capable of gelation at 37 °C in vitro and in vivo, and is cytocompatible with a variety of cell types and islet-like tissues in vitro. Overall, this study demonstrates the characterisation of a novel protocol for the decellularization and delipidization of human pancreatic tissue for the production of acellular ECM and ECM hydrogel suitable for cell culture and transplantation applications. We also report a list of 120 proteins present within the human pancreatic matrisome.


Archive | 2010

Reprogramming t cells and hematopoietic cells

Matthew E. Brown; Elizabeth Rondon Dominguez; Randy Learish; Emile Nuwaysir; Deepika Rajesh; Amanda Mack

Collaboration


Dive into the Matthew E. Brown's collaboration.

Top Co-Authors

Avatar

William J. Burlingham

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Deepika Rajesh

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Brian E. McIntosh

Morgridge Institute for Research

View shared research outputs
Top Co-Authors

Avatar

Daniel M. Tremmel

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Ian G. Norman

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jeremy A. Sullivan

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jon S. Odorico

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Sara Dutton Sackett

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Ying Zhou

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Arick C. Park

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge