Network


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

Hotspot


Dive into the research topics where Prasenjit Sarkar is active.

Publication


Featured researches published by Prasenjit Sarkar.


Analytical Chemistry | 2010

Direct comparison of stable isotope labeling by amino acids in cell culture and spectral counting for quantitative proteomics.

Timothy S. Collier; Prasenjit Sarkar; William L. Franck; Balaji M. Rao; Ralph A. Dean; David C. Muddiman

Numerous experimental strategies exist for relative protein quantification, one of the primary objectives of mass spectrometry based proteomics analysis. These strategies mostly involve the incorporation of a stable isotope label via either metabolic incorporation in cell or tissue culture (¹⁵N/¹⁴N metabolic labeling, stable isotope labeling by amino acids in cell culture (SILAC)), chemical derivatization (ICAT, iTRAQ, TMT), or enzymatically catalyzed incorporation (¹⁸O labeling). Also, these techniques can be cost or time prohibitive or not amenable to the biological system of interest (i.e., metabolic labeling of clinical samples, most animals, or fungi). This is the case with the quantification of fungal proteomes, which often require auxotroph mutants to fully metabolically label. Alternatively, label-free strategies for protein quantification such as using integrated ion abundance and spectral counting have been demonstrated for quantification affording over 2 orders of magnitude of dynamic range which is comparable to metabolic labeling strategies. Direct comparisons of these quantitative techniques are largely lacking in the literature but are highly warranted in order to evaluate the capabilities, limitations, and analytical variability of available quantitative strategies. Here, we present the direct comparison of SILAC to label-free quantification by spectral counting of an identical set of data from the bottom-up proteomic analysis of human embryonic stem cells, which are readily able to be quantified using both strategies, finding that both strategies result in a similar number of protein identifications. We also discuss necessary constraints for accurate quantification using spectral counting and assess the potential of this label-free strategy as a viable alternative for quantitative proteomics.


Journal of the American Society for Mass Spectrometry | 2010

Quantitative top-down proteomics of SILAC labeled human embryonic stem cells

Timothy S. Collier; Prasenjit Sarkar; Balaji M. Rao; David C. Muddiman

Human embryonic stem cells (hESCs) are self-renewing pluripotent cells with relevance to treatment of numerous medical conditions. However, a global understanding of the role of the hESC proteome in maintaining pluripotency or triggering differentiation is still largely lacking. The emergence of top-down proteomics has facilitated the identification and characterization of intact protein forms that are not readily apparent in bottom-up studies. Combined with metabolic labeling techniques such as stable isotope labeling by amino acids in cell culture (SILAC), quantitative comparison of intact protein expression under differing experimental conditions is possible. Herein, quantitative top-down proteomics of hESCs is demonstrated using the SILAC method and nano-flow reverse phase chromatography directly coupled to a linear-ion-trap Fourier transform ion cyclotron resonance mass spectrometer (nLC-LTQ-FT-ICR-MS). In this study, which to the best of our knowledge represents the first top-down analysis of hESCs, we have confidently identified 11 proteins by accurate intact mass, MS/MS, and amino acid counting facilitated by SILAC labeling. Although quantification is challenging due to the incorporation of multiple labeled amino acids (i.e., lysine and arginine) and arginine to proline conversion, we are able to quantitatively account for these phenomena using a mathematical model.


Rapid Communications in Mass Spectrometry | 2011

Comparison of stable-isotope labeling with amino acids in cell culture and spectral counting for relative quantification of protein expression.

Timothy S. Collier; Shan M. Randall; Prasenjit Sarkar; Balaji M. Rao; Ralph A. Dean; David C. Muddiman

Protein quantification is one of the principal goals of mass spectrometry (MS)-based proteomics, and many strategies exist to achieve it. Several approaches involve the incorporation of a stable-isotope label using either chemical derivatization, enzymatically catalyzed incorporation of (18)O, or metabolic labeling in a cell or tissue culture. These techniques can be cost or time prohibitive or not amenable to the biological system of interest. Label-free techniques including those utilizing integrated ion abundance and spectral counting offer an alternative to stable-isotope-based methodologies. Herein, we present the comparison of stable-isotope labeling of amino acids in cell culture (SILAC) with spectral counting for the quantification of human embryonic stem cells as they differentiate toward the trophectoderm at three time points. Our spectral counting experimental strategy resulted in the identification of 2641 protein groups across three time points with an average sequence coverage of 30.3%, of which 1837 could be quantified with more than five spectral counts. SILAC quantification was able to identify 1369 protein groups with an average coverage of 24.7%, of which 1027 could be quantified across all time points. Within this context we further explore the capacity of each strategy for proteome coverage, variation in quantification, and the relative sensitivity of each technique to the detection of change in relative protein expression.


Molecular & Cellular Proteomics | 2012

Targeted proteomics of the secretory pathway reveals the secretome of mouse embryonic fibroblasts and human embryonic stem cells.

Prasenjit Sarkar; Shan M. Randall; David C. Muddiman; Balaji M. Rao

Proteins endogenously secreted by human embryonic stem cells (hESCs) and those present in hESC culture medium are critical regulators of hESC self-renewal and differentiation. Current MS-based approaches for identifying secreted proteins rely predominantly on MS analysis of cell culture supernatants. Here we show that targeted proteomics of secretory pathway organelles is a powerful alternate approach for interrogating the cellular secretome. We have developed procedures to obtain subcellular fractions from mouse embryonic fibroblasts (MEFs) and hESCs that are enriched in secretory pathway organelles while ensuring retention of the secretory cargo. MS analysis of these fractions from hESCs cultured in MEF conditioned medium (MEF-CM) or MEFs exposed to hESC medium revealed 99 and 129 proteins putatively secreted by hESCs and MEFs, respectively. Of these, 53 and 62 proteins have been previously identified in cell culture supernatants of MEFs and hESCs, respectively, thus establishing the validity of our approach. Furthermore, 76 and 37 putatively secreted proteins identified in this study in MEFs and hESCs, respectively, have not been reported in previous MS analyses. The identification of low abundance secreted proteins via MS analysis of cell culture supernatants typically necessitates the use of altered culture conditions such as serum-free medium. However, an altered medium formulation might directly influence the cellular secretome. Indeed, we observed significant differences between the abundances of several secreted proteins in subcellular fractions isolated from hESCs cultured in MEF-CM and those exposed to unconditioned hESC medium for 24 h. In contrast, targeted proteomics of secretory pathway organelles does not require the use of customized media. We expect that our approach will be particularly valuable in two contexts highly relevant to hESC biology: obtaining a temporal snapshot of proteins secreted in response to a differentiation trigger, and identifying proteins secreted by cells that are isolated from a heterogeneous population.


Proteomics | 2012

The subcellular proteome of undifferentiated human embryonic stem cells

Prasenjit Sarkar; Timothy S. Collier; Shan M. Randall; David C. Muddiman; Balaji M. Rao

We have characterized the subcellular proteome of human embryonic stem cells (hESCs) through MS analysis of the membrane, cytosolic, and nuclear fractions, isolated from the same sample of undifferentiated hESCs. Strikingly, 74% of all proteins identified were detected in a single subcellular fraction; we also carried out immunofluorescence studies to validate the subcellular localization suggested by proteomic analysis, for a subset of proteins. Our approach resulted in deeper proteome coverage – peptides mapping to 893, 2475, and 1185 proteins were identified in the nuclear, cytosolic, and membrane fractions, respectively. Additionally, we used spectral counting to estimate the relative abundance of all cytosolic proteins. A large number of proteins relevant to hESC biology, including growth factor receptors, cell junction proteins, transcription factors, chromatin remodeling proteins, and histone modifying enzymes were identified. Our analysis shows that components of a large number of interacting signaling pathways are expressed in hESCs. Finally, we show that proteomic analysis of the endoplasmic reticulum (ER) and Golgi compartments is a powerful alternative approach to identify secreted proteins since these are synthesized in the ER and transit through the Golgi. Taken together, our results show that systematic subcellular proteomic analysis is a valuable tool for studying hESC biology.


Journal of Biological Chemistry | 2015

Activin/Nodal Signaling Switches the Terminal Fate of Human Embryonic Stem Cell-derived Trophoblasts

Prasenjit Sarkar; Shan M. Randall; Timothy S. Collier; Anthony Nero; Teal A. Russell; David C. Muddiman; Balaji M. Rao

Background: Specification of terminal fate in trophoblasts derived from human embryonic stem cells is not understood. Results: Inhibition of activin/nodal signaling triggers extravillous fate, but loss of inhibition causes syncytial fate. Conclusion: Activin/nodal signaling switches the terminal fate of trophoblasts. Significance: We provide a model system that allows for targeted derivation of extravillous trophoblasts and syncytiotrophoblasts. Human embryonic stem cells (hESCs) have been routinely treated with bone morphogenetic protein and/or inhibitors of activin/nodal signaling to obtain cells that express trophoblast markers. Trophoblasts can terminally differentiate to either extravillous trophoblasts or syncytiotrophoblasts. The signaling pathways that govern the terminal fate of these trophoblasts are not understood. We show that activin/nodal signaling switches the terminal fate of these hESC-derived trophoblasts. Inhibition of activin/nodal signaling leads to formation of extravillous trophoblast, whereas loss of activin/nodal inhibition leads to the formation of syncytiotrophoblasts. Also, the ability of hESCs to form bona fide trophoblasts has been intensely debated. We have examined hESC-derived trophoblasts in the light of stringent criteria that were proposed recently, such as hypomethylation of the ELF5-2b promoter region and down-regulation of HLA class I antigens. We report that trophoblasts that possess these properties can indeed be obtained from hESCs.


Biotechnology Journal | 2013

Trophoblast Differentiation of Human Embryonic Stem Cells.

Karthik Tiruthani; Prasenjit Sarkar; Balaji M. Rao

Molecular mechanisms regulating human trophoblast differentiation remain poorly understood due to difficulties in obtaining primary tissues from very early developmental stages in humans. Therefore, the use of human embryonic stem cells (hESCs) as a source for generating trophoblast tissues is of significant interest. Trophoblast‐like cells have been obtained through treatment of hESCs with bone morphogenetic protein (BMP) or inhibitors of activin/nodal/transforming growth factor‐β signaling, or through protocols involving formation of embryoid bodies (EBs); however, there is controversy over whether hESC‐derived cells are indeed analogous to true trophoblasts found in vivo. In this review, we provide an overview of previously described efforts to obtain trophoblasts from hESCs. We also discuss the merits and limitations of hESCs as a source of trophoblast derivatives.


Journal of Proteome Research | 2016

Identification of Epigenetic Factor Proteins Expressed in Human Embryonic Stem Cell-Derived Trophoblasts and in Human Placental Trophoblasts.

Prasenjit Sarkar; Adam Mischler; Shan M. Randall; Timothy S. Collier; Karen F. Dorman; Kim A. Boggess; David C. Muddiman; Balaji M. Rao

Human embryonic stem cells (hESCs) have been used to derive trophoblasts through differentiation in vitro. Intriguingly, mouse ESCs are prevented from differentiation to trophoblasts by certain epigenetic factor proteins such as Dnmt1, thus necessitating the study of epigenetic factor proteins during hESC differentiation to trophoblasts. We used stable isotope labeling by amino acids in cell culture and quantitative proteomics to study changes in the nuclear proteome during hESC differentiation to trophoblasts and identified changes in the expression of 30 epigenetic factor proteins. Importantly, the DNA methyltransferases DNMT1, DNMT3A, and DNMT3B were downregulated. Additionally, we hypothesized that nuclear proteomics of hESC-derived trophoblasts may be used for screening epigenetic factor proteins expressed by primary trophoblasts in human placental tissue. Accordingly, we conducted immunohistochemistry analysis of six epigenetic factor proteins identified from hESC-derived trophoblasts-DNMT1, DNMT3B, BAF155, BAF60A, BAF57, and ING5-in 6-9 week human placentas. Indeed, expression of these proteins was largely, though not fully, consistent with that observed in 6-9 week placental trophoblasts. Our results support the use of hESC-derived trophoblasts as a model for placental trophoblasts, which will enable further investigation of epigenetic factors involved in human trophoblast development.


Archive | 2011

Role of Signaling Pathways and Epigenetic Factors in Lineage Determination During Human Embryonic Stem Cell Differentiation

Prasenjit Sarkar; Balaji M. Rao

Human embryonic stem cells (hESCs) are culture-adapted cells that were originally derived from the inner cell mass (ICM) of the blastocyst-stage embryo [1]. HESCs are pluripotent cells that can be propagated indefinitely in culture, while retaining the in vivo properties of ICM cells; they can give rise to all tissues of the three germ layers (ectoderm, mesoderm and endoderm). Due to their pluripotency, hESCs have been the subject of intense research since they were initially isolated in 1998. HESCs can serve as model systems to study early human development, in addition to providing a potentially unlimited source of functional tissues for use in drug evaluation and regenerative medicine. Nevertheless, despite major advances, the exact molecular mechanisms that govern the self-renewal and differentiation of hESCs remain unclear. Indeed, a mechanistic understanding of the molecular processes regulating hESC fate can elucidate early events in human development and enable the development of protocols for efficient generation of functional tissues. Here we review the molecular mechanisms that regulate hESC fate; specifically, we focus on the role of signaling pathways and factors regulating epigenetic changes, in hESC self-renewal and lineage-specific differentiation. In hESCs, as in embryos, differentiation is triggered by developmental cues such as morphogens or cytokines that are present in the extracellular space. These morphogens or cytokines bind to their cognate plasma membrane-bound receptors and activate specific signaling pathways inside the cell. Activation of signaling pathways involves a sequence of phosphorylation events that eventually result in the regulation of specific transcription factors. These transcription factors, in turn, can recruit other co-factors and directly cause transcription of downstream genes. Furthermore, transcription factors can recruit histone modifying and chromatin remodeling enzymes to reshuffle the epigenetic structure, such that pluripotency genes become inaccessible for transcription and are repressed, whereas lineage-specific genes become accessible and are activated. This sequence of events finally leads to expression of lineage-specific proteins such as transcription factors and structural proteins, causing a morphological change in the cell. Also, pluripotency associated transcription factors and other pluripotency-associated genes are permanently repressed, thereby completing the process of differentiation. Thus, the process of differentiation is a


Critical Reviews in Biomedical Engineering | 2009

Molecular aspects of cardiac differentiation in embryonic stem cells.

Prasenjit Sarkar; Balaji M. Rao

Collaboration


Dive into the Prasenjit Sarkar's collaboration.

Top Co-Authors

Avatar

Balaji M. Rao

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

David C. Muddiman

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Timothy S. Collier

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Shan M. Randall

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Adam Mischler

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Ralph A. Dean

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Anthony Nero

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Karthik Tiruthani

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Teal A. Russell

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge