Network


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

Hotspot


Dive into the research topics where Divya Nag is active.

Publication


Featured researches published by Divya Nag.


Nature Biotechnology | 2011

An antibody against SSEA-5 glycan on human pluripotent stem cells enables removal of teratoma-forming cells

Chad Tang; Andrew S. Lee; Jens Peter Volkmer; Debashis Sahoo; Divya Nag; Adriane R. Mosley; Matthew A. Inlay; Reza Ardehali; Shawn L. Chavez; Renee A. Reijo Pera; B. Behr; Joseph C. Wu; Irving L. Weissman; Micha Drukker

An important risk in the clinical application of human pluripotent stem cells (hPSCs), including human embryonic and induced pluripotent stem cells (hESCs and hiPSCs), is teratoma formation by residual undifferentiated cells. We raised a monoclonal antibody against hESCs, designated anti–stage-specific embryonic antigen (SSEA)-5, which binds a previously unidentified antigen highly and specifically expressed on hPSCs—the H type-1 glycan. Separation based on SSEA-5 expression through fluorescence-activated cell sorting (FACS) greatly reduced teratoma-formation potential of heterogeneously differentiated cultures. To ensure complete removal of teratoma-forming cells, we identified additional pluripotency surface markers (PSMs) exhibiting a large dynamic expression range during differentiation: CD9, CD30, CD50, CD90 and CD200. Immunohistochemistry studies of human fetal tissues and bioinformatics analysis of a microarray database revealed that concurrent expression of these markers is both common and specific to hPSCs. Immunodepletion with antibodies against SSEA-5 and two additional PSMs completely removed teratoma-formation potential from incompletely differentiated hESC cultures.


Journal of the American College of Cardiology | 2012

Human Cardiac Progenitor Cells Engineered With Pim-I Kinase Enhance Myocardial Repair

Sadia Mohsin; Mohsin Khan; Haruhiro Toko; Brandi Bailey; Christopher T. Cottage; Kathleen Wallach; Divya Nag; Andrew Lee; Sailay Siddiqi; Feng Lan; Kimberlee Fischer; Natalie Gude; Pearl Quijada; Daniele Avitabile; Silvia Truffa; Brett Collins; Walter P. Dembitsky; Joseph C. Wu; Mark A. Sussman

OBJECTIVES The goal of this study was to demonstrate the enhancement of human cardiac progenitor cell (hCPC) reparative and regenerative potential by genetic modification for the treatment of myocardial infarction. BACKGROUND Regenerative potential of stem cells to repair acute infarction is limited. Improved hCPC survival, proliferation, and differentiation into functional myocardium will increase efficacy and advance translational implementation of cardiac regeneration. METHODS hCPCs isolated from the myocardium of heart failure patients undergoing left ventricular assist device implantation were engineered to express green fluorescent protein (hCPCe) or Pim-1-GFP (hCPCeP). Functional tests of hCPC regenerative potential were performed with immunocompromised mice by using intramyocardial adoptive transfer injection after infarction. Myocardial structure and function were monitored by echocardiographic and hemodynamic assessment for 20 weeks after delivery. hCPCe and hCPCeP expressing luciferase were observed by using bioluminescence imaging to noninvasively track persistence. RESULTS hCPCeP exhibited augmentation of reparative potential relative to hCPCe control cells, as shown by significantly increased proliferation coupled with amelioration of infarction injury and increased hemodynamic performance at 20 weeks post-transplantation. Concurrent with enhanced cardiac structure and function, hCPCeP demonstrated increased cellular engraftment and differentiation with improved vasculature and reduced infarct size. Enhanced persistence of hCPCeP versus hCPCe was revealed by bioluminescence imaging at up to 8 weeks post-delivery. CONCLUSIONS Genetic engineering of hCPCs with Pim-1 enhanced repair of damaged myocardium. Ex vivo gene delivery to modify stem cells has emerged as a viable option addressing current limitations in the field. This study demonstrates that efficacy of hCPCs from the failing myocardium can be safely and significantly enhanced through expression of Pim-1 kinase, setting the stage for use of engineered cells in pre-clinical settings.


Circulation | 2011

Novel MicroRNA Prosurvival Cocktail for Improving Engraftment and Function of Cardiac Progenitor Cell Transplantation

Shijun Hu; Mei Huang; Patricia K. Nguyen; Yongquan Gong; Zongjin Li; Fangjun Jia; Feng Lan; Junwei Liu; Divya Nag; Robert C. Robbins; Joseph C. Wu

Background— Although stem cell therapy has provided a promising treatment for myocardial infarction, the low survival of the transplanted cells in the infarcted myocardium is possibly a primary reason for failure of long-term improvement. Therefore, the development of novel prosurvival strategies to boost stem cell survival will be of significant benefit to this field. Methods and Results— Cardiac progenitor cells (CPCs) were isolated from transgenic mice, which constitutively express firefly luciferase and green fluorescent protein. The CPCs were transduced with individual lentivirus carrying the precursor of miR-21, miR-24, and miR-221, a cocktail of these 3 microRNA precursors, or green fluorescent protein as a control. After challenge in serum free medium, CPCs treated with the 3 microRNA cocktail showed significantly higher viability compared with untreated CPCs. After intramuscular and intramyocardial injections, in vivo bioluminescence imaging showed that microRNA cocktail-treated CPCs survived significantly longer after transplantation. After left anterior descending artery ligation, microRNA cocktail-treated CPCs boost the therapeutic efficacy in terms of functional recovery. Histological analysis confirmed increased myocardial wall thickness and CPC engraftment in the myocardium with the microRNA cocktail. Finally, we used bioinformatics analysis and experimental validation assays to show that Bim, a critical apoptotic activator, is an important target gene of the microRNA cocktail, which collectively can bind to the 3′UTR region of Bim and suppress its expression. Conclusions— We have demonstrated that a microRNA prosurvival cocktail (miR-21, miR-24, and miR-221) can improve the engraftment of transplanted cardiac progenitor cells and therapeutic efficacy for treatment of ischemic heart disease.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Clonal precursor of bone, cartilage, and hematopoietic niche stromal cells

Charles K. Chan; Paul Lindau; Wen Jiang; James Y. Chen; Lillian F. Zhang; Ching Cheng Chen; Jun Seita; Debashis Sahoo; Jae Beom Kim; Andrew Lee; Sujin Park; Divya Nag; Yongquan Gong; Subhash Kulkarni; Cynthia A. Luppen; Alexander A. Theologis; Derrick C. Wan; Anthony T. DeBoer; Eun Young Seo; Justin Vincent-Tompkins; Kyle M. Loh; Graham G. Walmsley; Daniel Kraft; Joseph C. Wu; Michael T. Longaker; Irving L. Weissman

Organs are composites of tissue types with diverse developmental origins, and they rely on distinct stem and progenitor cells to meet physiological demands for cellular production and homeostasis. How diverse stem cell activity is coordinated within organs is not well understood. Here we describe a lineage-restricted, self-renewing common skeletal progenitor (bone, cartilage, stromal progenitor; BCSP) isolated from limb bones and bone marrow tissue of fetal, neonatal, and adult mice. The BCSP clonally produces chondrocytes (cartilage-forming) and osteogenic (bone-forming) cells and at least three subsets of stromal cells that exhibit differential expression of cell surface markers, including CD105 (or endoglin), Thy1 [or CD90 (cluster of differentiation 90)], and 6C3 [ENPEP glutamyl aminopeptidase (aminopeptidase A)]. These three stromal subsets exhibit differential capacities to support hematopoietic (blood-forming) stem and progenitor cells. Although the 6C3-expressing subset demonstrates functional stem cell niche activity by maintaining primitive hematopoietic stem cell (HSC) renewal in vitro, the other stromal populations promote HSC differentiation to more committed lines of hematopoiesis, such as the B-cell lineage. Gene expression analysis and microscopic studies further reveal a microenvironment in which CD105-, Thy1-, and 6C3-expressing marrow stroma collaborate to provide cytokine signaling to HSCs and more committed hematopoietic progenitors. As a result, within the context of bone as a blood-forming organ, the BCSP plays a critical role in supporting hematopoiesis through its generation of diverse osteogenic and hematopoietic-promoting stroma, including HSC supportive 6C3(+) niche cells.


Journal of Biological Chemistry | 2011

Preclinical derivation and imaging of autologously transplanted canine induced pluripotent stem cells

Andrew S. Lee; Dan Xu; Jordan R. Plews; Patricia K. Nguyen; Divya Nag; Jennifer Lyons; Leng Han; Shijun Hu; Feng Lan; Junwei Liu; Mei Huang; Kazim H. Narsinh; Long Ct; Patricia E. de Almeida; Benjamin Levi; Nigel G. Kooreman; Charles D. Bangs; Cholawat Pacharinsak; Fumiaki Ikeno; Alan C. Yeung; Sanjiv S. Gambhir; Robert C. Robbins; Michael T. Longaker; Joseph C. Wu

Derivation of patient-specific induced pluripotent stem cells (iPSCs) opens a new avenue for future applications of regenerative medicine. However, before iPSCs can be used in a clinical setting, it is critical to validate their in vivo fate following autologous transplantation. Thus far, preclinical studies have been limited to small animals and have yet to be conducted in large animals that are physiologically more similar to humans. In this study, we report the first autologous transplantation of iPSCs in a large animal model through the generation of canine iPSCs (ciPSCs) from the canine adipose stromal cells and canine fibroblasts of adult mongrel dogs. We confirmed pluripotency of ciPSCs using the following techniques: (i) immunostaining and quantitative PCR for the presence of pluripotent and germ layer-specific markers in differentiated ciPSCs; (ii) microarray analysis that demonstrates similar gene expression profiles between ciPSCs and canine embryonic stem cells; (iii) teratoma formation assays; and (iv) karyotyping for genomic stability. Fate of ciPSCs autologously transplanted to the canine heart was tracked in vivo using clinical positron emission tomography, computed tomography, and magnetic resonance imaging. To demonstrate clinical potential of ciPSCs to treat models of injury, we generated endothelial cells (ciPSC-ECs) and used these cells to treat immunodeficient murine models of myocardial infarction and hindlimb ischemia.


Circulation | 2011

Double knockdown of prolyl hydroxylase and factor-inhibiting hypoxia-inducible factor with nonviral minicircle gene therapy enhances stem cell mobilization and angiogenesis after myocardial infarction.

Mei Huang; Patricia K. Nguyen; Fangjun Jia; Shijun Hu; Yongquan Gong; Patricia E. de Almeida; Wang L; Divya Nag; Mark A. Kay; Amato J. Giaccia; Robert C. Robbins; Joseph C. Wu

Background— Under normoxic conditions, hypoxia-inducible factor (HIF)-1&agr; is rapidly degraded by 2 hydroxylases: prolyl hydroxylase (PHD) and factor-inhibiting HIF-1 (FIH). Because HIF-1&agr; mediates the cardioprotective response to ischemic injury, its upregulation may be an effective therapeutic option for ischemic heart failure. Methods and Results— PHD and FIH were cloned from mouse embryonic stem cells. The best candidate short hairpin (sh) sequences for inhibiting PHD isoenzyme 2 and FIH were inserted into novel, nonviral, minicircle vectors. In vitro studies after cell transfection of mouse C2C12 myoblasts, HL-1 atrial myocytes, and c-kit+ cardiac progenitor cells demonstrated higher expression of angiogenesis factors in the double-knockdown group compared with the single-knockdown and short hairpin scramble control groups. To confirm in vitro data, shRNA minicircle vectors were injected intramyocardially after left anterior descending coronary artery ligation in adult FVB mice (n=60). Functional studies using MRI, echocardiography, and pressure-volume loops showed greater improvement in cardiac function in the double-knockdown group. To assess mechanisms of this functional recovery, we performed a cell trafficking experiment, which demonstrated significantly greater recruitment of bone marrow cells to the ischemic myocardium in the double-knockdown group. Fluorescence-activated cell sorting showed significantly higher activation of endogenous c-kit+ cardiac progenitor cells. Immunostaining showed increased neovascularization and decreased apoptosis in areas of injured myocardium. Finally, western blots and laser-capture microdissection analysis confirmed upregulation of HIF-1&agr; protein and angiogenesis genes, respectively. Conclusions— We demonstrated that HIF-1&agr; upregulation by double knockdown of PHD and FIH synergistically increases stem cell mobilization and myocardial angiogenesis, leading to improved cardiac function.


Circulation-cardiovascular Imaging | 2012

Early stem cell engraftment predicts late cardiac functional recovery: preclinical insights from molecular imaging.

Junwei Liu; Kazim H. Narsinh; Feng Lan; Wang L; Patricia K. Nguyen; Shijun Hu; Andrew Lee; Leng Han; Yongquan Gong; Mei Huang; Divya Nag; Jarrett Rosenberg; Alexandra Chouldechova; Robert C. Robbins; Joseph C. Wu

Background— Human cardiac progenitor cells have demonstrated great potential for myocardial repair in small and large animals, but robust methods for longitudinal assessment of their engraftment in humans is not yet readily available. In this study, we sought to optimize and evaluate the use of positron emission tomography (PET) reporter gene imaging for monitoring human cardiac progenitor cell (hCPC) transplantation in a mouse model of myocardial infarction. Methods and Results— hCPCs were isolated and expanded from human myocardial samples and stably transduced with herpes simplex virus thymidine kinase (TK) PET reporter gene. Thymidine kinase-expressing hCPCs were characterized in vitro and transplanted into murine myocardial infarction models (n=57). Cardiac echocardiographic, magnetic resonance imaging and pressure-volume loop analyses revealed improvement in left ventricular contractile function 2 weeks after transplant (hCPC versus phosphate-buffered saline, P<0.03). Noninvasive PET imaging was used to track hCPC fate over a 4-week time period, demonstrating a substantial decline in surviving cells. Importantly, early cell engraftment as assessed by PET was found to predict subsequent functional improvement, implying a “dose–effect” relationship. We isolated the transplanted cells from recipient myocardium by laser capture microdissection for in vivo transcriptome analysis. Our results provide direct evidence that hCPCs augment cardiac function after their transplantation into ischemic myocardium through paracrine secretion of growth factors. Conclusions— PET reporter gene imaging can provide important diagnostic and prognostic information regarding the ultimate success of hCPC treatment for myocardial infarction.


Circulation | 2012

Safe Genetic Modification of Cardiac Stem Cells Using a Site-Specific Integration Technique

Feng Lan; Junwei Liu; Kazim H. Narsinh; Shijun Hu; Leng Han; Andrew S. Lee; Marisa Karow; Patricia K. Nguyen; Divya Nag; Michele P. Calos; Robert C. Robbins; Joseph C. Wu

Background— Human cardiac progenitor cells (hCPCs) are a promising cell source for regenerative repair after myocardial infarction. Exploitation of their full therapeutic potential may require stable genetic modification of the cells ex vivo. Safe genetic engineering of stem cells, using facile methods for site-specific integration of transgenes into known genomic contexts, would significantly enhance the overall safety and efficacy of cellular therapy in a variety of clinical contexts. Methods and Results— We used the phiC31 site-specific recombinase to achieve targeted integration of a triple fusion reporter gene into a known chromosomal context in hCPCs and human endothelial cells. Stable expression of the reporter gene from its unique chromosomal integration site resulted in no discernible genomic instability or adverse changes in cell phenotype. Namely, phiC31-modified hCPCs were unchanged in their differentiation propensity, cellular proliferative rate, and global gene expression profile when compared with unaltered control hCPCs. Expression of the triple fusion reporter gene enabled multimodal assessment of cell fate in vitro and in vivo using fluorescence microscopy, bioluminescence imaging, and positron emission tomography. Intramyocardial transplantation of genetically modified hCPCs resulted in significant improvement in myocardial function 2 weeks after cell delivery, as assessed by echocardiography (P=0.002) and MRI (P=0.001). We also demonstrated the feasibility and therapeutic efficacy of genetically modifying differentiated human endothelial cells, which enhanced hind limb perfusion (P<0.05 at day 7 and 14 after transplantation) on laser Doppler imaging. Conclusions— The phiC31 integrase genomic modification system is a safe, efficient tool to enable site-specific integration of reporter transgenes in progenitor and differentiated cell types.


Advanced Drug Delivery Reviews | 2010

Methods to Assess Stem Cell Lineage, Fate and Function

Patricia K. Nguyen; Divya Nag; Joseph C. Wu

Stem cell therapy has the potential to regenerate injured tissue. For stem cells to achieve their full therapeutic potential, stem cells must differentiate into the target cell, reach the site of injury, survive, and engraft. To fully characterize these cells, evaluation of cell morphology, lineage specific markers, cell specific function, and gene expression must be performed. To monitor survival and engraftment, cell fate imaging is vital. Only then can organ specific function be evaluated to determine the effectiveness of therapy. In this review, we will discuss methods for evaluating the function of transplanted cells for restoring the heart, nervous system, and pancreas. We will also highlight the specific challenges facing these potential therapeutic areas.


Journal of Nuclear Cardiology | 2011

Sex Differences in the Diagnostic Evaluation of Coronary Artery Disease

Patricia K. Nguyen; Divya Nag; Joseph C. Wu

Although sex differences in coronary heart disease (CHD) have long been recognized, many of the recommendations for the management of female patients continue to be identical to male patients. Given the paucity of sex-specific data in basic science and clinical studies, however, defining unique diagnostic and therapeutic strategies for women remains problematic for scientists and clinicians. For instance, women represent only 38% of subjects in previously NIH-funded cardiovascular studies (1). Previous studies and clinical trials have also included inadequate numbers of women. Finally, only 25% of previous cardiovascular clinical trials have reported sex-specific results (2). Recently, researchers have been encouraged to report sex differences in basic and clinical studies. Much of the impetus originates from data indicating that more women die of cardiovascular disease (CVD) than men (3). This disparity in mortality may signal the need for sex-specific guidelines for the diagnosis of CHD. In this review, we will discuss sex differences in the clinical manifestations and outcome of CHD, the limitations of current approaches for the management of female patients, and the potential strategies to improve the evaluation of CHD in women.

Collaboration


Dive into the Divya Nag'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

Leng Han

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge