Bio

Honors & Awards


  • Horizon Award, Department of Defense, USA (2016)

Professional Education


  • B of Medicine and B of Surgery, Bundelkhand University (2005)
  • Master of Technology, Indian Institute of Technology, Kanpur (2008)
  • Doctor of Philosophy, S.U.N.Y. State University at Buffalo (2015)

Stanford Advisors


Research & Scholarship

Current Research and Scholarly Interests


Molecular regulation of stem cell plasticity in development and disease
Tissue Engineering and Regenerative Medicine

Publications

All Publications


  • Reprogramming Postnatal Human Epidermal Keratinocytes Toward Functional Neural Crest Fates STEM CELLS Bajpai, V. K., Kerosuo, L., Tseropoulos, G., Cummings, K. A., Wang, X., Lei, P., Liu, B., Liu, S., Popescu, G. K., Bronner, M. E., Andreadis, S. T. 2017; 35 (5): 1402-1415

    Abstract

    During development, neural crest (NC) cells are induced by signaling events at the neural plate border of all vertebrate embryos. Initially arising within the central nervous system, NC cells subsequently undergo an epithelial to mesenchymal transition to migrate into the periphery, where they differentiate into diverse cell types. Here we provide evidence that postnatal human epidermal keratinocytes (KC), in response to fibroblast growth factor 2 and insulin like growth factor 1 signals, can be reprogrammed toward a NC fate. Genome-wide transcriptome analyses show that keratinocyte-derived NC cells are similar to those derived from human embryonic stem cells. Moreover, they give rise in vitro and in vivo to NC derivatives such as peripheral neurons, melanocytes, Schwann cells and mesenchymal cells (osteocytes, chondrocytes, adipocytes, and smooth muscle cells). By demonstrating that human keratin-14+ KC can form NC cells, even from clones of single cells, our results have important implications in stem cell biology and regenerative medicine. Stem Cells 2017;35:1402-1415.

    View details for DOI 10.1002/stem.2583

    View details for Web of Science ID 000400017200024

    View details for PubMedID 28142205

  • NANOG Reverses the Myogenic Differentiation Potential of Senescent Stem Cells by Restoring ACTIN Filamentous Organization and SRF-Dependent Gene Expression STEM CELLS Mistriotis, P., Bajpai, V. K., Wang, X., Rong, N., Shahini, A., Asmani, M., Liang, M., Wang, J., Lei, P., Liu, S., Zhao, R., Andreadis, S. T. 2017; 35 (1): 207-221

    Abstract

    Cellular senescence as a result of organismal aging or progeroid diseases leads to stem cell pool exhaustion hindering tissue regeneration and contributing to the progression of age related disorders. Here we discovered that ectopic expression of the pluripotent factor NANOG in senescent or progeroid myogenic progenitors reversed cellular aging and restored completely the ability to generate contractile force. To elicit its effects, NANOG enabled reactivation of the ROCK and Transforming Growth Factor (TGF)-β pathways-both of which were impaired in senescent cells-leading to ACTIN polymerization, MRTF-A translocation into the nucleus and serum response factor (SRF)-dependent myogenic gene expression. Collectively our data reveal that cellular senescence can be reversed and provide a novel strategy to regain the lost function of aged stem cells without reprogramming to the pluripotent state. Stem Cells 2017;35:207-221.

    View details for DOI 10.1002/stem.2452

    View details for Web of Science ID 000391966700022

    View details for PubMedID 27350449

  • Flow induced adherens junction remodeling driven by cytoskeletal forces. Experimental cell research Verma, D., Bajpai, V. K., Ye, N., Maneshi, M. M., Jetta, D., Andreadis, S. T., Sachs, F., Hua, S. Z. 2017

    Abstract

    Adherens junctions (AJs) are a key structural component for tissue organization and function. Under fluid shear stress, AJs exhibit dynamic assembly/disassembly, but how shear stress couples to AJs is unclear. In MDCK cells we measured simultaneously the forces in cytoskeletal α-actinin and the density and length of AJs using a genetically coded optical force sensor, actinin-sstFRET, and fluorescently labeled E-cadherin (E-cad). We found that shear stress of 0.74dyn/cm(2) for 3h significantly enhanced E-cad expression at cell-cell contacts and this phenomenon has two phases. The initial formation of segregated AJ plaques coincided with a decrease in cytoskeletal tension, but an increase in tension was necessary for expansion of the plaques and the formation of continuous AJs in the later phase. The changes in cytoskeletal tension and reorganization appear to be an upstream process in response to flow since it occurred in both wild type and dominant negative E-cad cells. Disruption of F-actin with a Rho-ROCK inhibitor eliminated AJ growth under flow. These results delineate the shear stress transduction paths in cultured cells, which helps to understand pathology of a range of diseases that involve dysfunction of E-cadherin.

    View details for DOI 10.1016/j.yexcr.2017.08.009

    View details for PubMedID 28803065

  • Heart Regeneration with Engineered Myocardial Tissue ANNUAL REVIEW OF BIOMEDICAL ENGINEERING, VOL 16 Coulombe, K. L., Bajpai, V. K., Andreadis, S. T., Murry, C. E. 2014; 16: 1-28

    Abstract

    Heart disease is the leading cause of morbidity and mortality worldwide, and regenerative therapies that replace damaged myocardium could benefit millions of patients annually. The many cell types in the heart, including cardiomyocytes, endothelial cells, vascular smooth muscle cells, pericytes, and cardiac fibroblasts, communicate via intercellular signaling and modulate each other's function. Although much progress has been made in generating cells of the cardiovascular lineage from human pluripotent stem cells, a major challenge now is creating the tissue architecture to integrate a microvascular circulation and afferent arterioles into such an engineered tissue. Recent advances in cardiac and vascular tissue engineering will move us closer to the goal of generating functionally mature tissue. Using the biology of the myocardium as the foundation for designing engineered tissue and addressing the challenges to implantation and integration, we can bridge the gap from bench to bedside for a clinically tractable engineered cardiac tissue.

    View details for DOI 10.1146/annurev-bioeng-071812-152344

    View details for Web of Science ID 000348433000001

    View details for PubMedID 24819474

    View details for PubMedCentralID PMC4213953

  • Functional vascular smooth muscle cells derived from human induced pluripotent stem cells via mesenchymal stem cell intermediates CARDIOVASCULAR RESEARCH Bajpai, V. K., Mistriotis, P., Loh, Y., Daley, G. Q., Andreadis, S. T. 2012; 96 (3): 391-400

    Abstract

    Smooth muscle cells (SMC) play an important role in vascular homeostasis and disease. Although adult mesenchymal stem cells (MSC) have been used as a source of contractile SMC, they suffer from limited proliferation potential and culture senescence, particularly when originating from older donors. By comparison, human induced pluripotent stem cells (hiPSC) can provide an unlimited source of functional SMC for autologous cell-based therapies and for creating models of vascular disease. Our goal was to develop an efficient strategy to derive functional, contractile SMC from hiPSC.We developed a robust, stage-wise, feeder-free strategy for hiPSC differentiation into functional SMC through an intermediate stage of multipotent MSC, which could be coaxed to differentiate into fat, bone, cartilage, and muscle. At this stage, the cells were highly proliferative and displayed higher clonogenic potential and reduced senescence when compared with parental hair follicle mesenchymal stem cells. In addition, when exposed to differentiation medium, the myogenic proteins such as α-smooth muscle actin, calponin, and myosin heavy chain were significantly upregulated and displayed robust fibrillar organization, suggesting the development of a contractile phenotype. Indeed, tissue constructs prepared from these cells exhibited high levels of contractility in response to receptor- and non-receptor-mediated agonists.We developed an efficient stage-wise strategy that enabled hiPSC differentiation into contractile SMC through an intermediate population of clonogenic and multipotent MSC. The high yield of MSC and SMC derivation suggests that our strategy may facilitate an acquisition of the large numbers of cells required for regenerative medicine or for studying vascular disease pathophysiology.

    View details for DOI 10.1093/cvr/cvs253

    View details for Web of Science ID 000311306800010

    View details for PubMedID 22941255

    View details for PubMedCentralID PMC3584971

  • Stem Cell Sources for Vascular Tissue Engineering and Regeneration TISSUE ENGINEERING PART B-REVIEWS Bajpai, V. K., Andreadis, S. T. 2012; 18 (5): 405-425

    Abstract

    This review focuses on the stem cell sources with the potential to be used in vascular tissue engineering and to promote vascular regeneration. The first clinical studies using tissue-engineered vascular grafts are already under way, supporting the potential of this technology in the treatment of cardiovascular and other diseases. Despite progress in engineering biomaterials with the appropriate mechanical properties and biological cues as well as bioreactors for generating the correct tissue microenvironment, the source of cells that make up the vascular tissues remains a major challenge for tissue engineers and physicians. Mature cells from the tissue of origin may be difficult to obtain and suffer from limited proliferative capacity, which may further decline as a function of donor age. On the other hand, multipotent and pluripotent stem cells have great potential to provide large numbers of autologous cells with a great differentiation capacity. Here, we discuss the adult multipotent as well as embryonic and induced pluripotent stem cells, their differentiation potential toward vascular lineages, and their use in engineering functional and implantable vascular tissues. We also discuss the associated challenges that need to be addressed in order to facilitate the transition of this technology from the bench to the bedside.

    View details for DOI 10.1089/ten.teb.2011.0264

    View details for Web of Science ID 000309516500006

    View details for PubMedID 22571595

    View details for PubMedCentralID PMC3458622

  • Clonal multipotency and effect of long-term in vitro expansion on differentiation potential of human hair follicle derived mesenchymal stem cells STEM CELL RESEARCH Bajpai, V. K., Mistriotis, P., Andreadis, S. T. 2012; 8 (1): 74-84

    Abstract

    Hair follicle harbors a rich stem cell pool with mesenchymal lineage differentiation potential. Although previous studies with rodent cells demonstrated that hair follicle sheath and papilla cells possess multi-lineage differentiation potential, human hair follicle derived mesenchymal stem cells (hHF-MSCs) have not been characterized in detail in terms of their multipotency. In addition, it is not clear whether these cells are true stem cells that can differentiate along multiple lineages or whether they represent a collection of progenitor cells with restricted differentiation potential. Here we report that hHF-MSCs are highly proliferative cells that can be maintained in culture for ~45 population doublings before they start to show signs of cellular senescence. Under appropriate culture conditions, hHF-MSCs differentiated along the myogenic, osteogenic, adipogenic and chondrogenic lineages, as demonstrated by kinetic gene expression profiling and functional assays. Interestingly, the differentiation potential decreased with time in culture in a lineage-specific manner. Specifically, myogenesis and chondrogenesis showed a moderate decrease over time; osteogenesis was maximum at intermediate passages and adipogenesis was highly sensitive to long-term culture and was diminished at late passages. Finally, hHF-MSCs were clonally multipotent as the majority of hHF-MSCs clones (73%) demonstrated bi- or tri-lineage differentiation potential. These results suggest that hHF-MSCs may present as an alternative source of easily accessible, autologous stem cells for tissue engineering and regenerative medicine.

    View details for DOI 10.1016/j.scr.2011.07.003

    View details for Web of Science ID 000300129000007

    View details for PubMedID 22099022

    View details for PubMedCentralID PMC3222855

  • Cation-pi interaction: to stack or to spread MOLECULAR PHYSICS Mishra, B. K., Bajpai, V. K., Ramanathan, V., Gadre, S. R., Sathyamurthy, N. 2008; 106 (12-13): 1557-1566