Network


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

Hotspot


Dive into the research topics where Nutan Prasain is active.

Publication


Featured researches published by Nutan Prasain.


Blood | 2011

Hematopoietic stem/progenitor cells, generation of induced pluripotent stem cells, and isolation of endothelial progenitors from 21- to 23.5-year cryopreserved cord blood

Hal E. Broxmeyer; Man Ryul Lee; Giao Hangoc; Scott Cooper; Nutan Prasain; Young June Kim; Coleen Mallett; Zhaohui Ye; Scott R. Witting; Kenneth Cornetta; Linzhao Cheng; Mervin C. Yoder

Cryopreservation of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) is crucial for cord blood (CB) banking and transplantation. We evaluated recovery of functional HPC cryopreserved as mononuclear or unseparated cells for up to 23.5 years compared with prefreeze values of the same CB units. Highly efficient recovery (80%-100%) was apparent for granulocyte-macrophage and multipotential hematopoietic progenitors, although some collections had reproducible low recovery. Proliferative potential, response to multiple cytokines, and replating of HPC colonies was extensive. CD34(+) cells isolated from CB cryopreserved for up to 21 years had long-term (≥ 6 month) engrafting capability in primary and secondary immunodeficient mice reflecting recovery of long-term repopulating, self-renewing HSCs. We recovered functionally responsive CD4(+) and CD8(+) T lymphocytes, generated induced pluripotent stem (iPS) cells with differentiation representing all 3 germ cell lineages in vitro and in vivo, and detected high proliferative endothelial colony forming cells, results of relevance to CB biology and banking.


Nature Biotechnology | 2014

Differentiation of human pluripotent stem cells to cells similar to cord-blood endothelial colony–forming cells

Nutan Prasain; Man Ryul Lee; Sasidhar Vemula; Jonathan Luke Meador; Momoko Yoshimoto; Michael J. Ferkowicz; Alexa Fett; Manav Gupta; Brian M. Rapp; Mohammad Reza Saadatzadeh; Michael Ginsberg; Olivier Elemento; Younghee Lee; Sherry L. Voytik-Harbin; Hyung Min Chung; Ki Sung Hong; Emma Reid; Christina O'Neill; Reinhold Medina; Alan W. Stitt; Michael P. Murphy; Shahin Rafii; Hal E. Broxmeyer; Mervin C. Yoder

The ability to differentiate human pluripotent stem cells into endothelial cells with properties of cord-blood endothelial colony–forming cells (CB-ECFCs) may enable the derivation of clinically relevant numbers of highly proliferative blood vessel–forming cells to restore endothelial function in patients with vascular disease. We describe a protocol to convert human induced pluripotent stem cells (hiPSCs) or embryonic stem cells (hESCs) into cells similar to CB-ECFCs at an efficiency of >108 ECFCs produced from each starting pluripotent stem cell. The CB-ECFC-like cells display a stable endothelial phenotype with high clonal proliferative potential and the capacity to form human vessels in mice and to repair the ischemic mouse retina and limb, and they lack teratoma formation potential. We identify Neuropilin-1 (NRP-1)-mediated activation of KDR signaling through VEGF165 as a critical mechanism for the emergence and maintenance of CB-ECFC-like cells.


Stem Cells | 2013

Epigenetic Regulation of Nanog by MiR-302 Cluster-MBD2 Completes Induced Pluripotent Stem Cell Reprogramming

Man Ryul Lee; Nutan Prasain; Hee Don Chae; Young June Kim; Charlie Mantel; Mervin C. Yoder; Hal E. Broxmeyer

While most somatic cells undergoing induced pluripotent stem (iPS) cell reprogramming with Yamanaka factors accumulate at stable partially reprogrammed stages, the molecular mechanisms required to achieve full reprogramming are unknown. MicroRNAs (miRNAs) fine‐tune mRNA translation and are implicated in reprogramming, but miRNA functional targets critical for complete iPS cell reprogramming remain elusive. We identified methyl‐DNA binding domain protein 2 (MBD2) as an epigenetic suppressor, blocking full reprogramming of somatic to iPS cells through direct binding to NANOG promoter elements preventing transcriptional activation. When we overexpressed miR‐302 cluster we observed a significant increase in conversion of partial to fully reprogrammed iPS cells by suppressing MBD2 expression, thereby increasing NANOG expression. Thus, expression of exogenous miR‐302 cluster (without miR‐367) is efficient in attaining a fully reprogrammed iPS state in partially reprogrammed cells by relieving MBD2‐mediated inhibition of NANOG expression. Our studies provide a direct molecular mechanism involved in generating complete human iPS cell reprogramming to study disease pathogenesis, drug screening, and for potential cell‐based therapies. STEM CELLS 2013;31:666–681


American Journal of Physiology-lung Cellular and Molecular Physiology | 2014

The Pseudomonas aeruginosa exoenzyme Y impairs endothelial cell proliferation and vascular repair following lung injury

Trevor C. Stevens; Cristhiaan D. Ochoa; K. Adam Morrow; Matthew Robson; Nutan Prasain; Chun Zhou; Diego F. Alvarez; Dara W. Frank; Ron Balczon; Troy Stevens

Exoenzyme Y (ExoY) is a Pseudomonas aeruginosa toxin that is introduced into host cells through the type 3 secretion system (T3SS). Once inside the host cell cytoplasm, ExoY generates cyclic nucleotides that cause tau phosphorylation and microtubule breakdown. Microtubule breakdown causes interendothelial cell gap formation and tissue edema. Although ExoY transiently induces interendothelial cell gap formation, it remains unclear whether ExoY prevents repair of the endothelial cell barrier. Here, we test the hypothesis that ExoY intoxication impairs recovery of the endothelial cell barrier following gap formation, decreasing migration, proliferation, and lung repair. Pulmonary microvascular endothelial cells (PMVECs) were infected with P. aeruginosa strains for 6 h, including one possessing an active ExoY (PA103 exoUexoT::Tc pUCPexoY; ExoY(+)), one with an inactive ExoY (PA103ΔexoUexoT::Tc pUCPexoY(K81M); ExoY(K81M)), and one that lacks PcrV required for a functional T3SS (ΔPcrV). ExoY(+) induced interendothelial cell gaps, whereas ExoY(K81M) and ΔPcrV did not promote gap formation. Following gap formation, bacteria were removed and endothelial cell repair was examined. PMVECs were unable to repair gaps even 3-5 days after infection. Serum-stimulated growth was greatly diminished following ExoY intoxication. Intratracheal inoculation of ExoY(+) and ExoY(K81M) caused severe pneumonia and acute lung injury. However, whereas the pulmonary endothelial cell barrier was functionally improved 1 wk following ExoY(K81M) infection, pulmonary endothelium was unable to restrict the hyperpermeability response to elevated hydrostatic pressure following ExoY(+) infection. In conclusion, ExoY is an edema factor that chronically impairs endothelial cell barrier integrity following lung injury.


Current Opinion in Organ Transplantation | 2010

Inducible pluripotent stem cells: Not quite ready for prime time?

Reiesha D. Robbins; Nutan Prasain; Bernhard Maier; Mervin C. Yoder; Raghavendra G. Mirmira

Purpose of reviewInducible pluripotent stem (iPS) cells derived from somatic cells represent a novel renewable source of tissue precursors. The potential of iPS cells is considered to be at least equivalent to that of human embryonic stem cells, facilitating the treatment or cure of diseases such as diabetes mellitus, spinal cord injuries, cardiovascular disease, and neurodegenerative diseases, but with the potential added benefit of evading the adaptive immune response that otherwise limits allogeneic cell-based therapies. This review discusses recent advances in pluripotency induction and the use of iPS cells to produce differentiated cells, while highlighting roadblocks to the widespread use of this technology in the clinical arena. Recent findingsWhereas ethical and safety issues surrounding the use of human embryonic stem cells for the treatment of disease continue to be debated, use of iPS cells may be viewed as a more widely acceptable compromise. Since the first descriptions of inducible pluripotency from somatic cells, multiple laboratories have collectively made tremendous strides both in developing alternative, more clinically acceptable, induction strategies and in demonstrating the proof-of-principle that iPS cells can be differentiated into a variety of cell types to reverse mouse models of human disease. SummaryAlthough the prospect of using patient-specific iPS cells has much appeal from an ethical and immunologic perspective, the limitations of the technology from the standpoint of reprogramming efficiency and therapeutic safety necessitate much more in-depth research before the initiation of human clinical trials.


Journal of Visualized Experiments | 2012

Phenotypic and Functional Characterization of Endothelial Colony Forming Cells Derived from Human Umbilical Cord Blood

Nutan Prasain; J. Luke Meador; Mervin C. Yoder

Longstanding views of new blood vessel formation via angiogenesis, vasculogenesis, and arteriogenesis have been recently reviewed. The presence of circulating endothelial progenitor cells (EPCs) were first identified in adult human peripheral blood by Asahara et al. in 1997 bringing an infusion of new hypotheses and strategies for vascular regeneration and repair. EPCs are rare but normal components of circulating blood that home to sites of blood vessel formation or vascular remodeling, and facilitate either postnatal vasculogenesis, angiogenesis, or arteriogenesis largely via paracrine stimulation of existing vessel wall derived cells. No specific marker to identify an EPC has been identified, and at present the state of the field is to understand that numerous cell types including proangiogenic hematopoietic stem and progenitor cells, circulating angiogenic cells, Tie2+ monocytes, myeloid progenitor cells, tumor associated macrophages, and M2 activated macrophages participate in stimulating the angiogenic process in a variety of preclinical animal model systems and in human subjects in numerous disease states. Endothelial colony forming cells (ECFCs) are rare circulating viable endothelial cells characterized by robust clonal proliferative potential, secondary and tertiary colony forming ability upon replating, and ability to form intrinsic in vivo vessels upon transplantation into immunodeficient mice. While ECFCs have been successfully isolated from the peripheral blood of healthy adult subjects, umbilical cord blood (CB) of healthy newborn infants, and vessel wall of numerous human arterial and venous vessels. CB possesses the highest frequency of ECFCs that display the most robust clonal proliferative potential and form durable and functional blood vessels in vivo. While the derivation of ECFC from adult peripheral blood has been presented, here we describe the methodologies for the derivation, cloning, expansion, and in vitro as well as in vivo characterization of ECFCs from the human umbilical CB.


PLOS ONE | 2013

Pseudomonas aeruginosa Exotoxin Y-Mediated Tau Hyperphosphorylation Impairs Microtubule Assembly in Pulmonary Microvascular Endothelial Cells

Ronald Balczon; Nutan Prasain; Cristhiaan D. Ochoa; Jason Prater; Bing Zhu; Mikhail Alexeyev; Sarah Sayner; Dara W. Frank; Troy Stevens

Pseudomonas aeruginosa uses a type III secretion system to introduce the adenylyl and guanylyl cyclase exotoxin Y (ExoY) into the cytoplasm of endothelial cells. ExoY induces Tau hyperphosphorylation and insolubility, microtubule breakdown, barrier disruption and edema, although the mechanism(s) responsible for microtubule breakdown remain poorly understood. Here we investigated both microtubule behavior and centrosome activity to test the hypothesis that ExoY disrupts microtubule dynamics. Fluorescence microscopy determined that infected pulmonary microvascular endothelial cells contained fewer microtubules than control cells, and further studies demonstrated that the microtubule-associated protein Tau was hyperphosphorylated following infection and dissociated from microtubules. Disassembly/reassembly studies determined that microtubule assembly was disrupted in infected cells, with no detectable effects on either microtubule disassembly or microtubule nucleation by centrosomes. This effect of ExoY on microtubules was abolished when the cAMP-dependent kinase phosphorylation site (Ser-214) on Tau was mutated to a non-phosphorylatable form. These studies identify Tau in microvascular endothelial cells as the target of ExoY in control of microtubule architecture following pulmonary infection by Pseudomonas aeruginosa and demonstrate that phosphorylation of tau following infection decreases microtubule assembly.


Stem Cells and Development | 2017

Metabolome Profiling of Partial and Fully Reprogrammed Induced Pluripotent Stem Cells

Soon Jung Park; Sang A. Lee; Nutan Prasain; Daekyeong Bae; Hyunsu Kang; Taewon Ha; Jong Soo Kim; Ki Sung Hong; Charlie Mantel; Sung Hwan Moon; Hal E. Broxmeyer; Man Ryul Lee

Acquisition of proper metabolomic fate is required to convert somatic cells toward fully reprogrammed pluripotent stem cells. The majority of induced pluripotent stem cells (iPSCs) are partially reprogrammed and have a transcriptome different from that of the pluripotent stem cells. The metabolomic profile and mitochondrial metabolic functions required to achieve full reprogramming of somatic cells to iPSC status have not yet been elucidated. Clarification of the metabolites underlying reprogramming mechanisms should enable further optimization to enhance the efficiency of obtaining fully reprogrammed iPSCs. In this study, we characterized the metabolites of human fully reprogrammed iPSCs, partially reprogrammed iPSCs, and embryonic stem cells (ESCs). Using capillary electrophoresis time-of-flight mass spectrometry-based metabolomics, we found that 89% of analyzed metabolites were similarly expressed in fully reprogrammed iPSCs and human ESCs (hESCs), whereas partially reprogrammed iPSCs shared only 74% similarly expressed metabolites with hESCs. Metabolomic profiling analysis suggested that converting mitochondrial respiration to glycolytic flux is critical for reprogramming of somatic cells into fully reprogrammed iPSCs. This characterization of metabolic reprogramming in iPSCs may enable the development of new reprogramming parameters for enhancing the generation of fully reprogrammed human iPSCs.


Biotechnology Journal | 2017

iPSC‐Derived Vascular Cell Spheroids as Building Blocks for Scaffold‐Free Biofabrication

Leni Moldovan; April L. Barnard; Chang Hyun Gil; Yang Lin; Maria B. Grant; Mervin C. Yoder; Nutan Prasain; Nicanor I. Moldovan


Stem cell reports | 2017

Epigenetic Activation of Pro-angiogenic Signaling Pathways in Human Endothelial Progenitors Increases Vasculogenesis

Sylvain Fraineau; Carmen G. Palii; Brian McNeill; Morten Ritso; William C. Shelley; Nutan Prasain; Alphonse Chu; Elodie Vion; Kristy Rieck; Sharmin Nilufar; Theodore J. Perkins; Michael A. Rudnicki; David S. Allan; Mervin C. Yoder; Erik J. Suuronen; Marjorie Brand

Collaboration


Dive into the Nutan Prasain's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Troy Stevens

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dara W. Frank

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Diego F. Alvarez

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mikhail Alexeyev

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ron Balczon

University of South Alabama

View shared research outputs
Researchain Logo
Decentralizing Knowledge