Network


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

Hotspot


Dive into the research topics where Brenden W. Smith is active.

Publication


Featured researches published by Brenden W. Smith.


Stem Cells | 2010

Generation of Transgene-Free Lung Disease-Specific Human Induced Pluripotent Stem Cells Using a Single Excisable Lentiviral Stem Cell Cassette

Aba Somers; Jyh-Chang Jean; Cesar A. Sommer; Amel Omari; Christopher C. Ford; Jason A. Mills; Lei Ying; Andreia Gianotti Sommer; Jenny M. Jean; Brenden W. Smith; Robert Lafyatis; Marie-France Demierre; Daniel J. Weiss; Deborah L. French; Paul Gadue; George J. Murphy; Gustavo Mostoslavsky; Darrell N. Kotton

The development of methods to achieve efficient reprogramming of human cells while avoiding the permanent presence of reprogramming transgenes represents a critical step toward the use of induced pluripotent stem cells (iPSC) for clinical purposes, such as disease modeling or reconstituting therapies. Although several methods exist for generating iPSC free of reprogramming transgenes from mouse cells or neonatal normal human tissues, a sufficiently efficient reprogramming system is still needed to achieve the widespread derivation of disease‐specific iPSC from humans with inherited or degenerative diseases. Here, we report the use of a humanized version of a single lentiviral “stem cell cassette” vector to accomplish efficient reprogramming of normal or diseased skin fibroblasts obtained from humans of virtually any age. Simultaneous transfer of either three or four reprogramming factors into human target cells using this single vector allows derivation of human iPSC containing a single excisable viral integration that on removal generates human iPSC free of integrated transgenes. As a proof of principle, here we apply this strategy to generate >100 lung disease‐specific iPSC lines from individuals with a variety of diseases affecting the epithelial, endothelial, or interstitial compartments of the lung, including cystic fibrosis, α‐1 antitrypsin deficiency‐related emphysema, scleroderma, and sickle‐cell disease. Moreover, we demonstrate that human iPSC generated with this approach have the ability to robustly differentiate into definitive endoderm in vitro, the developmental precursor tissue of lung epithelia. STEM CELLS 2010;28:1728–1740


Blood | 2013

The aryl hydrocarbon receptor directs hematopoietic progenitor cell expansion and differentiation

Brenden W. Smith; Sarah S. Rozelle; Amy Leung; Jessalyn Ubellacker; Ashley Parks; Shirley K. Nah; Deborah L. French; Paul Gadue; Stefano Monti; David H.K. Chui; Martin H. Steinberg; Alan D. Michelson; Roger Théberge; Mark E. McComb; Catherine E. Costello; Darrell N. Kotton; Gustavo Mostoslavsky; David H. Sherr; George J. Murphy

The evolutionarily conserved aryl hydrocarbon receptor (AhR) has been studied for its role in environmental chemical-induced toxicity. However, recent studies have demonstrated that the AhR may regulate the hematopoietic and immune systems during development in a cell-specific manner. These results, together with the absence of an in vitro model system enabling production of large numbers of primary human hematopoietic progenitor cells (HPs) capable of differentiating into megakaryocyte- and erythroid-lineage cells, motivated us to determine if AhR modulation could facilitate both progenitor cell expansion and megakaryocyte and erythroid cell differentiation. Using a novel, pluripotent stem cell-based, chemically-defined, serum and feeder cell-free culture system, we show that the AhR is expressed in HPs and that, remarkably, AhR activation drives an unprecedented expansion of HPs, megakaryocyte-lineage cells, and erythroid-lineage cells. Further AhR modulation within rapidly expanding progenitor cell populations directs cell fate, with chronic AhR agonism permissive to erythroid differentiation and acute antagonism favoring megakaryocyte specification. These results highlight the development of a new Good Manufacturing Practice-compliant platform for generating virtually unlimited numbers of human HPs with which to scrutinize red blood cell and platelet development, including the assessment of the role of the AhR critical cell fate decisions during hematopoiesis.


Journal of Visualized Experiments | 2012

Generation of human induced pluripotent stem cells from peripheral blood using the STEMCCA lentiviral vector

Andreia Gianotti Sommer; Sarah S. Rozelle; Spencer K. Sullivan; Jason A. Mills; Seonmi Park; Brenden W. Smith; Amulya Iyer; Deborah L. French; Darrell N. Kotton; Paul Gadue; George J. Murphy; Gustavo Mostoslavsky

Through the ectopic expression of four transcription factors, Oct4, Klf4, Sox2 and cMyc, human somatic cells can be converted to a pluripotent state, generating so-called induced pluripotent stem cells (iPSCs)(1-4). Patient-specific iPSCs lack the ethical concerns that surround embryonic stem cells (ESCs) and would bypass possible immune rejection. Thus, iPSCs have attracted considerable attention for disease modeling studies, the screening of pharmacological compounds, and regenerative therapies(5). We have shown the generation of transgene-free human iPSCs from patients with different lung diseases using a single excisable polycistronic lentiviral Stem Cell Cassette (STEMCCA) encoding the Yamanaka factors(6). These iPSC lines were generated from skin fibroblasts, the most common cell type used for reprogramming. Normally, obtaining fibroblasts requires a skin punch biopsy followed by expansion of the cells in culture for a few passages. Importantly, a number of groups have reported the reprogramming of human peripheral blood cells into iPSCs(7-9). In one study, a Tet inducible version of the STEMCCA vector was employed(9), which required the blood cells to be simultaneously infected with a constitutively active lentivirus encoding the reverse tetracycline transactivator. In contrast to fibroblasts, peripheral blood cells can be collected via minimally invasive procedures, greatly reducing the discomfort and distress of the patient. A simple and effective protocol for reprogramming blood cells using a constitutive single excisable vector may accelerate the application of iPSC technology by making it accessible to a broader research community. Furthermore, reprogramming of peripheral blood cells allows for the generation of iPSCs from individuals in which skin biopsies should be avoided (i.e. aberrant scarring) or due to pre-existing disease conditions preventing access to punch biopsies. Here we demonstrate a protocol for the generation of human iPSCs from peripheral blood mononuclear cells (PBMCs) using a single floxed-excisable lentiviral vector constitutively expressing the 4 factors. Freshly collected or thawed PBMCs are expanded for 9 days as described(10,11) in medium containing ascorbic acid, SCF, IGF-1, IL-3 and EPO before being transduced with the STEMCCA lentivirus. Cells are then plated onto MEFs and ESC-like colonies can be visualized two weeks after infection. Finally, selected clones are expanded and tested for the expression of the pluripotency markers SSEA-4, Tra-1-60 and Tra-1-81. This protocol is simple, robust and highly consistent, providing a reliable methodology for the generation of human iPSCs from readily accessible 4 ml of blood.


Current Opinion in Hematology | 2014

Stem cells, megakaryocytes, and platelets.

Brenden W. Smith; George J. Murphy

Purpose of reviewStem cells are an important tool for the study of ex-vivo models of megakaryopoiesis and the production of functional platelets. In this manuscript, we review the optimization of megakaryocyte and platelet differentiation and discuss the mechanistic studies and disease models that have incorporated stem cell technologies. Recent findingsMechanisms of cytoskeletal regulation and signal transduction have revealed insights into hierarchical dynamics of hematopoiesis, highlighting the close relationship between hematopoietic stem cells and cells of the megakaryocyte lineage. Platelet disorders have been successfully modeled and genetically corrected, and differentiation strategies have been optimized to the extent that utilizing stem cell-derived platelets for cellular therapy is feasible. SummaryStudies that utilize stem cells for the efficient derivation of megakaryocytes and platelets have played a role in uncovering novel molecular mechanisms of megakaryopoiesis, modeling and correcting relevant diseases, and differentiating platelets that are functional and scalable for translation into the clinic. Efforts to derive megakaryocytes and platelets from pluripotent stem cells foster the opportunity of a revolutionary cellular therapy for the treatment of multiple platelet-associated diseases.


Stem Cells International | 2016

Genome Editing of the CYP1A1 Locus in iPSCs as a Platform to Map AHR Expression throughout Human Development

Brenden W. Smith; Elizabeth A. Stanford; David H. Sherr; George J. Murphy

The aryl hydrocarbon receptor (AHR) is a ligand activated transcription factor that increases the expression of detoxifying enzymes upon ligand stimulation. Recent studies now suggest that novel endogenous roles of the AHR exist throughout development. In an effort to create an optimized model system for the study of AHR signaling in several cellular lineages, we have employed a CRISPR/CAS9 genome editing strategy in induced pluripotent stem cells (iPSCs) to incorporate a reporter cassette at the transcription start site of one of its canonical targets, cytochrome P450 1A1 (CYP1A1). This cell line faithfully reports on CYP1A1 expression, with luciferase levels as its functional readout, when treated with an endogenous AHR ligand (FICZ) at escalating doses. iPSC-derived fibroblast-like cells respond to acute exposure to environmental and endogenous AHR ligands, and iPSC-derived hepatocytes increase CYP1A1 in a similar manner to primary hepatocytes. This cell line is an important innovation that can be used to map AHR activity in discrete cellular subsets throughout developmental ontogeny. As further endogenous ligands are proposed, this line can be used to screen for safety and efficacy and can report on the ability of small molecules to regulate critical cellular processes by modulating the activity of the AHR.


BMC Biology | 2016

The role of the aryl hydrocarbon receptor in the development of cells with the molecular and functional characteristics of cancer stem-like cells

Elizabeth A. Stanford; Zhongyan Wang; Olga Novikov; Francesca Mulas; Esther Landesman-Bollag; Stefano Monti; Brenden W. Smith; David C. Seldin; George J. Murphy; David H. Sherr


Archive | 2016

Additional file 1: Additional Figure S1. of The role of the aryl hydrocarbon receptor in the development of cells with the molecular and functional characteristics of cancer stem-like cells

Elizabeth A. Stanford; Zhongyan Wang; Olga Novikov; Francesca Mulas; Esther Landesman-Bollag; Stefano Monti; Brenden W. Smith; David C. Seldin; George J. Murphy; David H. Sherr


Archive | 2016

The role of the aryl hydrocarbon receptor in the development of cells with the molecular and functional characteristics of

Elizabeth A. Stanford; Zhongyan Wang; Olga Novikov; Francesca Mulas; Esther Landesman-Bollag; Stefano Monti; Brenden W. Smith; David C. Seldin; George J. Murphy; David H. Sherr


Archive | 2013

Production de globules rouges et de plaquettes à partir de cellules souches

George J. Murphy; David H. Sherr; Sarah S. Rozelle; Brenden W. Smith


Blood | 2012

Induced Pluripotent Stem Cell Modeling of Sickle Cell Anemia

Sarah S. Rozelle; Brenden W. Smith; Efthymia Melista; Ehimen C. Aneni; Paola Sebastiani; Clinton T. Baldwin; Abdulrahman Alsultan; David H.K. Chui; Gustavo Mostoslavsky; Martin H. Steinberg; George J. Murphy

Collaboration


Dive into the Brenden W. Smith's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Deborah L. French

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul Gadue

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge