Network


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

Hotspot


Dive into the research topics where Kevin G. Chen is active.

Publication


Featured researches published by Kevin G. Chen.


Nature Biotechnology | 2007

Characterization of human embryonic stem cell lines by the International Stem Cell Initiative

Oluseun Adewumi; Behrouz Aflatoonian; Lars Ährlund-Richter; Michal Amit; Peter W. Andrews; Gemma Beighton; Paul Bello; Nissim Benvenisty; Lorraine S. Berry; Simon Bevan; Barak Blum; Justin Brooking; Kevin G. Chen; Andre Choo; Gary A. Churchill; Marie Corbel; Ivan Damjanov; John S Draper; Petr Dvorak; Katarina Emanuelsson; Roland A. Fleck; Angela Ford; Karin Gertow; Marina Gertsenstein; Paul J. Gokhale; Rebecca S. Hamilton; Alex Hampl; Lyn Healy; Outi Hovatta; Johan Hyllner

The International Stem Cell Initiative characterized 59 human embryonic stem cell lines from 17 laboratories worldwide. Despite diverse genotypes and different techniques used for derivation and maintenance, all lines exhibited similar expression patterns for several markers of human embryonic stem cells. They expressed the glycolipid antigens SSEA3 and SSEA4, the keratan sulfate antigens TRA-1-60, TRA-1-81, GCTM2 and GCT343, and the protein antigens CD9, Thy1 (also known as CD90), tissue-nonspecific alkaline phosphatase and class 1 HLA, as well as the strongly developmentally regulated genes NANOG, POU5F1 (formerly known as OCT4), TDGF1, DNMT3B, GABRB3 and GDF3. Nevertheless, the lines were not identical: differences in expression of several lineage markers were evident, and several imprinted genes showed generally similar allele-specific expression patterns, but some gene-dependent variation was observed. Also, some female lines expressed readily detectable levels of XIST whereas others did not. No significant contamination of the lines with mycoplasma, bacteria or cytopathic viruses was detected.


Cell Stem Cell | 2014

Human Pluripotent Stem Cell Culture: Considerations for Maintenance, Expansion, and Therapeutics

Kevin G. Chen; Barbara S. Mallon; Ronald D. G. McKay; Pamela Gehron Robey

Human pluripotent stem cells (hPSCs) provide powerful resources for application in regenerative medicine and pharmaceutical development. In the past decade, various methods have been developed for large-scale hPSC culture that rely on combined use of multiple growth components, including media containing various growth factors, extracellular matrices, 3D environmental cues, and modes of multicellular association. In this Protocol Review, we dissect these growth components by comparing cell culture methods and identifying the benefits and pitfalls associated with each one. We further provide criteria, considerations, and suggestions to achieve optimal cell growth for hPSC expansion, differentiation, and use in future therapeutic applications.


Pigment Cell & Melanoma Research | 2009

Involvement of ABC transporters in melanogenesis and the development of multidrug resistance of melanoma

Kevin G. Chen; Julio C. Valencia; Jean-Pierre Gillet; Vincent J. Hearing; Michael M. Gottesman

Because melanomas are intrinsically resistant to conventional radiotherapy and chemotherapy, many alternative treatment approaches have been developed such as biochemotherapy and immunotherapy. The most common cause of multidrug resistance (MDR) in human cancers is the expression and function of one or more ATP‐binding cassette (ABC) transporters that efflux anticancer drugs from cells. Melanoma cells express a group of ABC transporters (such as ABCA9, ABCB1, ABCB5, ABCB8, ABCC1, ABCC2, and ABCD1) that may be associated with the resistance of melanoma cells to a broad range of anticancer drugs and/or of melanocytes to toxic melanin intermediates and metabolites. In this review, we propose a model (termed the ABC‐M model) in which the intrinsic MDR of melanoma cells is at least in part because of the transporter systems that may also play a critical role in reducing the cytotoxicity of the melanogenic pathway in melanocytes. The ABC‐M model suggests molecular strategies to reverse MDR function in the context of the melanogenic pathway, which could open therapeutic avenues towards the ultimate goal of circumventing clinical MDR in patients with melanoma.


Journal of Cell Science | 2006

Sorting of Pmel17 to melanosomes through the plasma membrane by AP1 and AP2: evidence for the polarized nature of melanocytes

Julio C. Valencia; Hidenori Watabe; An Chi; Francois Rouzaud; Kevin G. Chen; Wilfred D. Vieira; Kaoruko Takahashi; Yuji Yamaguchi; Werner Berens; Kunio Nagashima; Jeffrey Shabanowitz; Donald F. Hunt; Ettore Appella; Vincent J. Hearing

Adaptor proteins (AP) play important roles in the sorting of proteins from the trans-Golgi network, but how they function in the sorting of various melanosome-specific proteins such as Pmel17, an essential structural component of melanosomes, in melanocytes is unknown. We characterized the processing and trafficking of Pmel17 via adaptor protein complexes within melanocytic cells. Proteomics analysis detected Pmel17, AP1 and AP2, but not AP3 or AP4 in early melanosomes. Real-time PCR, immunolabeling and tissue in-situ hybridization confirmed the coexpression of AP1 isoforms μ1A and μ1B (expressed only in polarized cells) in melanocytes and keratinocytes, but expression of μ1B is missing in some melanoma cell lines. Transfection with AP1 isoforms (μ1A or μ1B) showed two distinct distribution patterns that involved Pmel17, and only μ1B was able to restore the sorting of Pmel17 to the plasma membrane in cells lacking μ1B expression. Finally, we established that expression of μ1B is regulated physiologically in melanocytes by UV radiation or DKK1. These results show that Pmel17 is sorted to melanosomes by various intracellular routes, directly or indirectly through the plasma membrane, and the presence of basolateral elements in melanocytes suggests their polarized nature.


Journal of Cellular Physiology | 2005

Trafficking and localization of platinum complexes in cisplatin-resistant cell lines monitored by fluorescence-labeled platinum.

Xing-Jie Liang; Ding-Wu Shen; Kevin G. Chen; Stephen Wincovitch; Susan Garfield; Michael M. Gottesman

Cisplatin is a chemotherapeutic agent commonly used in the treatment of a wide variety of malignant tumors. Resistance to cisplatin represents a major obstacle to effective cancer therapy because clinically significant levels of resistance quickly emerge after treatment. Based on previous studies indicating abnormal plasma membrane protein trafficking in cisplatin‐resistant (CP‐r) cells, Fluorescence (Alexa Fluor)‐labeled cisplatin was used to determine whether this defect altered the trafficking and localization of cisplatin by comparing drug sensitive KB‐3‐1 and KB‐CP‐r cells. Alexa Fluor–cisplatin was readily internalized and localized throughout the KB‐3‐1 cells, but overall fluorescence decreased in KB‐CP‐r cells, as detected by flow cytometry (FACS) and confocal microscopy. Only punctate cytoplasmic staining was observed in KB‐CP‐r cells with less fluorescence observed in the nucleus. Colocalization experiments with a Golgi‐selective stain indicate the involvement of Golgi‐like vesicles in initial intracellular processing of Alexa Fluor conjugated cisplatin complexes. As detected using an antibody to Alexa Fluor–cisplatin, cisplatin complex‐binding proteins (CCBPs) were reduced in membrane fractions of single‐step cisplatin‐resistant KB‐CP.5 cells, and increased in the cytoplasm of KB‐CP.5 cells compared to KB‐3‐1 cells. CCBPs localized to lower density fractions in KB‐CP.5 cells than in KB‐3‐1 cells as determined by iodixanol gradient centrifugation. In summary, inappropriate trafficking of CCBPs might explain resistance to cisplatin in cultured cancer cells, presumably because membrane binding proteins for cisplatin are not properly located on the cell surface in these cells, but are instead trapped in low density vesicles within the cytoplasm.


Journal of the National Cancer Institute | 2009

Influence of Melanosome Dynamics on Melanoma Drug Sensitivity

Kevin G. Chen; Richard D. Leapman; Guofeng Zhang; Barry Lai; Julio C. Valencia; Carol O. Cardarelli; Wilfred D. Vieira; Vincent J. Hearing; Michael M. Gottesman

BACKGROUND Malignant melanomas are intrinsically resistant to many conventional treatments, such as radiation and chemotherapy, for reasons that are poorly understood. Here we propose and test a model that explains drug resistance or sensitivity in terms of melanosome dynamics. METHODS The growth and sensitivity to cisplatin of MNT-1 cells, which are melanotic and enriched with mature stage III and IV melanosomes, and SK-MEL-28 cells, which have only immature stage I and II melanosomes, were compared using clonogenic assays. Differences in pigmentation, melanosome stages, melanosome number, and cellular structures in different cell lines in response to various treatments were examined by electron microscopy. The relative numbers of melanosomes of different stages were compared after treatment with 1-phenyl-2-thiourea. The relationship between drug transporter function and endogenous melanogenic toxicity was assessed by treating cells with the cyclosporin analog PSC-833 and by assessing vacuole formation and cell growth inhibition. All statistical tests were two-sided. RESULTS Endogenous melanogenic cytotoxicity, produced by damaged melanosomes, resulted in pronounced cell growth inhibition in MNT-1 cells compared with amelanotic SK-MEL-28 cells. The sensitivity to CDDP of MNT-1 cells was 3.8-fold higher than that of SK-MEL-28 cells (mean IC(50) for SK-MEL-28 and MNT-1 = 2.13 microM and 0.56 microM, respectively; difference = 1.57 microM, 95% confidence interval = 1.45 to 1.69; P = .0017). After treatment with 6.7 microM CDDP for 72 hours, the number of stage II-III melanosomes in surviving MNT-1 cells was 6.8-fold that of untreated cells. Modulation of MNT-1 cells to earlier-stage (II, II-III, III) melanosomes by treatment with the tyrosinase inhibitor 1-phenyl-2-thiourea dramatically increased CDDP resistance. Furthermore, PSC-833 principally suppressed MNT-1 melanotic cell growth via an elevation of autophagosome-like vacuolar structures, possibly by inhibiting melanosome membrane transporters. CONCLUSIONS Melanosome dynamics (including their biogenesis, density, status, and structural integrity) regulate the drug resistance of melanoma cells. Manipulation of melanosome functions may be an effective way to enhance the therapeutic activity of anticancer drugs against melanoma.


Journal of Biological Chemistry | 2007

Sialylated Core 1 O-Glycans Influence the Sorting of Pmel17/gp100 and Determine Its Capacity to Form Fibrils

Julio C. Valencia; Francois Rouzaud; Sylvain Julien; Kevin G. Chen; Thierry Passeron; Yuji Yamaguchi; Mones Abu-Asab; Maria Tsokos; Gertrude E. Costin; Hiroshi Yamaguchi; Lisa M. Miller Jenkins; Kunio Nagashima; Ettore Appella; Vincent J. Hearing

Pmel17 is a melanocyte/melanoma-specific protein that is essential for the maturation of melanosomes to form mature, fibrillar, and pigmented organelles. Recently, we reported that the less glycosylated form of Pmel17 (termed iPmel17) is sorted via the plasma membrane in a manner distinct from mature Pmel17 (termed mPmel17), which is sorted directly to melanosomes. To clarify the mechanism(s) underlying the distinct processing and sorting of Pmel17, we generated a highly specific antibody (termed αPEP25h) against an epitope within the repeat domain of Pmel17 that is sensitive to changes in O-glycosylation. αPEP25h recognizes only iPmel17 and allows analysis of the processing and sorting of iPmel17 when compared with αPEP13h, an antibody that recognizes both iPmel17 and mPmel17. Our novel findings using αPEP25h demonstrate that iPmel17 differs from mPmel17 not only in its sensitivity to endoglycosidase H, but also in the content of core 1 O-glycans modified with sialic acid. This evidence reveals that iPmel17 is glycosylated differently in the Golgi and that it is sorted through the secretory pathway. Analysis of Pmel17 processing in glycosylation-deficient mutant cells reveals that Pmel17 lacking the correct addition of sialic acid and galactose loses the ability to form fibrils. Furthermore, we show that addition of sialic acid affects the stability and sorting of Pmel17 and reduces pigmentation. Alterations in sialyltransferase activity and substrates differ between normal and transformed melanocytes and may represent a critical change during malignant transformation.


The FASEB Journal | 2006

Regulation of constitutive and UVR-induced skin pigmentation by melanocortin 1 receptor isoforms

Francois Rouzaud; Gertrude-E. Costin; Yuji Yamaguchi; Julio C. Valencia; Werner Berens; Kevin G. Chen; Toshihiko Hoashi; Markus Böhm; Zalfa A. Abdel-Malek; Vincent J. Hearing

Melanin synthesized by epidermal melanocytes protects the skin against UVR‐induced DNA damage and skin cancer. Exposure to UVR increases the synthesis of the photoprotective eumelanin on activation of MC1R, a melanoma susceptibility gene. We studied the expression of MC1R under UVR and α‐MSH stimulation in skin of different ethnic origins and in melanocytes of various pigmentary levels. This study identifies and characterizes a novel MC1R isoform (MC1R350) generated by alternative splicing of the classically known MC1R (MC1R317). We demonstrate that the melanin content of melanocytes shows a significant positive correlation with MC1R317 levels but correlates inversely with the amount of MC1R350, suggesting that this latter isoform could act as a negative regulator of melanin synthesis. We confirmed that hypothesis by showing that while MC1R317 signaling significantly increases the expression of MITF and tyrosinase, two key factors in the melanin synthesis pathway, MC1R350 dramatically hampers their expression. In the skin, we show that UVR does not increase MC1R350 expression but does significantly increase MC1R317. Taken together, our results strongly suggest that MC1R350 acts as a negative regulator of skin pigmentation and demonstrate for the first time that MC1R isoform‐specific expression is closely related to skin pigmentation and photoprotection.—Rouzaud, F., Costin, G‐E., Yamaguchi, Y., Valencia, J. C., Berens, W. F., Chen, K. G., Hoashi, T., Böhm, M., Abdel‐Malek, Z. A., Hearing, V. J. Regulation of constitutive and UVR‐induced skin pigmentation by melanocortin 1 receptor isoforms. FASEB J. 20, E1338–E1350 (2006)


Stem Cells | 2012

Regulation and expression of the ATP-binding cassette transporter ABCG2 in human embryonic stem cells.

Raji Padmanabhan; Kevin G. Chen; Jean-Pierre Gillet; Misty Handley; Barbara S. Mallon; Rebecca S. Hamilton; Kye-Yoon Park; Sudhir Varma; Michele G. Mehaffey; Pamela Gehron Robey; Ronald D. G. McKay; Michael M. Gottesman

The expression and function of several multidrug transporters (including ABCB1 and ABCG2) have been studied in human cancer cells and in mouse and human adult stem cells. However, the expression of ABCG2 in human embryonic stem cells (hESCs) remains unclear. Limited and contradictory results in the literature from two research groups have raised questions regarding its expression and function. In this study, we used quantitative real‐time PCR, Northern blots, whole genome RNA sequencing, Western blots, and immunofluorescence microscopy to study ABCG2 expression in hESCs. We found that full‐length ABCG2 mRNA transcripts are expressed in undifferentiated hESC lines. However, ABCG2 protein was undetectable even under embryoid body differentiation or cytotoxic drug induction. Moreover, surface ABCG2 protein was coexpressed with the differentiation marker stage‐specific embryonic antigen‐1 of hESCs, following constant BMP‐4 signaling at days 4 and 6. This expression was tightly correlated with the downregulation of two microRNAs (miRNAs) (i.e., hsa‐miR‐519c and hsa‐miR‐520h). Transfection of miRNA mimics and inhibitors of these two miRNAs confirmed their direct involvement in the regulation ABCG2 translation. Our findings clarify the controversy regarding the expression of the ABCG2 gene and also provide new insights into translational control of the expression of membrane transporter mRNAs by miRNAs in hESCs. STEM Cells2012;30:2175–2187


Stem Cell Research | 2012

Non-colony type monolayer culture of human embryonic stem cells.

Kevin G. Chen; Barbara S. Mallon; Rebecca S. Hamilton; Olga A. Kozhich; Kye-Yoon Park; Daniel J. Hoeppner; Pamela Gehron Robey; Ronald D. G. McKay

Regenerative medicine, relying on human embryonic stem cell (hESC) technology, opens promising new avenues for therapy of many severe diseases. However, this approach is restricted by limited production of the desired cells due to the refractory properties of hESC growth in vitro. It is further hindered by insufficient control of cellular stress, growth rates, and heterogeneous cellular states under current culture conditions. In this study, we report a novel cell culture method based on a non-colony type monolayer (NCM) growth. Human ESCs under NCM remain pluripotent as determined by teratoma assays and sustain the potential to differentiate into three germ layers. This NCM culture has been shown to homogenize cellular states, precisely control growth rates, significantly increase cell production, and enhance hESC recovery from cryopreservation without compromising chromosomal integrity. This culture system is simple, robust, scalable, and suitable for high-throughput screening and drug discovery.

Collaboration


Dive into the Kevin G. Chen's collaboration.

Top Co-Authors

Avatar

Michael M. Gottesman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Pamela Gehron Robey

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Barbara S. Mallon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Julio C. Valencia

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Vincent J. Hearing

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Rebecca S. Hamilton

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Francois Rouzaud

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ronald D. G. McKay

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Kory R. Johnson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kye-Yoon Park

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge