Network


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

Hotspot


Dive into the research topics where Chulho Kang is active.

Publication


Featured researches published by Chulho Kang.


Journal of Clinical Investigation | 2003

Inhibition of adipogenesis and development of glucose intolerance by soluble preadipocyte factor–1 (Pref-1)

Kichoon Lee; Josep A. Villena; Yang Soo Moon; Kee-Hong Kim; Sunjoo Lee; Chulho Kang; Hei Sook Sul

Preadipocyte factor-1 (Pref-1) is a transmembrane protein highly expressed in preadipocytes. Pref-1 expression is, however, completely abolished in adipocytes. The extracellular domain of Pref-1 undergoes two proteolytic cleavage events that generate 50 and 25 kDa soluble products. To understand the function of Pref-1, we generated transgenic mice that express the full ectodomain corresponding to the large cleavage product of Pref-1 fused to human immunoglobulin-gamma constant region. Mice expressing the Pref-1/hFc transgene in adipose tissue, driven by the adipocyte fatty acid-binding protein (aP2, also known as aFABP) promoter, showed a substantial decrease in total fat pad weight. Moreover, adipose tissue from transgenic mice showed reduced expression of adipocyte markers and adipocyte-secreted factors, including leptin and adiponectin, whereas the preadipocyte marker Pref-1 was increased. Pref-1 transgenic mice with a substantial, but not complete, loss of adipose tissue exhibited hypertriglyceridemia, impaired glucose tolerance, and decreased insulin sensitivity. Mice expressing the Pref-1/hFc transgene exclusively in liver under the control of the albumin promoter also showed a decrease in adipose mass and adipocyte marker expression, suggesting an endocrine mode of action of Pref-1. These findings demonstrate the inhibition of adipogenesis by Pref-1 in vivo and the resulting impairment of adipocyte function that leads to the development of metabolic abnormalities.


Science | 2013

Aire-Dependent Thymic Development of Tumor-Associated Regulatory T Cells

Sven Malchow; Daniel S. Leventhal; Saki Nishi; Benjamin I. Fischer; Lynn Shen; Gladell P. Paner; Ayelet S. Amit; Chulho Kang; Jenna E. Geddes; James P. Allison; Nicholas D. Socci; Peter A. Savage

On the Origin of Tumor Tregs The tumor microenvironment is often seeded with regulatory T cells (Tregs), which inhibit antitumor immunity. Using mice with genetically driven prostate cancer, Malchow et al. (p. 1219; see the Perspective by Joshi and Jacks) found a population of Tregs that were enriched in the prostate of tumor-bearing mice. Surprisingly, these cells were also present in female mice and were found to be specific, not for a tumor-specific antigen, but rather for an antigen normally expressed in the prostate. Prostate antigen–specific Tregs arose in the thymus and their selection was dependent on Aire, a protein that drives the expression of tissue-specific antigens in the thymus. Thus, Tregs that seed tumors likely arise in the thymus, are not necessarily tumor-specific, and are recruited and/or expand in an organ when a tumor arises. Prostate tumors in mice recruit thymus-derived regulatory T cells that are specific for tissue autoantigens. [Also see Perspective by Joshi and Jacks] Despite considerable interest in the modulation of tumor-associated Foxp3+ regulatory T cells (Tregs) for therapeutic benefit, little is known about the developmental origins of these cells and the nature of the antigens that they recognize. We identified an endogenous population of antigen-specific Tregs (termed MJ23 Tregs) found recurrently enriched in the tumors of mice with oncogene-driven prostate cancer. MJ23 Tregs were not reactive to a tumor-specific antigen but instead recognized a prostate-associated antigen that was present in tumor-free mice. MJ23 Tregs underwent autoimmune regulator (Aire)–dependent thymic development in both male and female mice. Thus, Aire-mediated expression of peripheral tissue antigens drives the thymic development of a subset of organ-specific Tregs, which are likely coopted by tumors developing within the associated organ.


Nature Medicine | 2009

AdPLA ablation increases lipolysis and prevents obesity induced by high-fat feeding or leptin deficiency

Kathy Jaworski; Maryam Ahmadian; Robin E. Duncan; Eszter Sarkadi-Nagy; Krista A. Varady; Marc K. Hellerstein; Hui Young Lee; Varman T. Samuel; Gerald I. Shulman; Kee Hong Kim; Sarah De Val; Chulho Kang; Hei Sook Sul

A main function of white adipose tissue is to release fatty acids from stored triacylglycerol for other tissues to use as an energy source. Whereas endocrine regulation of lipolysis has been extensively studied, autocrine and paracrine regulation is not well understood. Here we describe the role of the newly identified major adipocyte phospholipase A2, AdPLA (encoded by Pla2g16, also called HREV107), in the regulation of lipolysis and adiposity. AdPLA-null mice have a markedly higher rate of lipolysis owing to increased cyclic AMP levels arising from the marked reduction in the amount of adipose prostaglandin E2 that binds the Gαi-coupled receptor, EP3. AdPLA-null mice have markedly reduced adipose tissue mass and triglyceride content but normal adipogenesis. They also have higher energy expenditure with increased fatty acid oxidation within adipocytes. AdPLA-deficient ob/ob mice remain hyperphagic but lean, with increased energy expenditure, yet have ectopic triglyceride storage and insulin resistance. AdPLA is a major regulator of adipocyte lipolysis and is crucial for the development of obesity.


Journal of Experimental Medicine | 2004

Essential Role of Survivin, an Inhibitor of Apoptosis Protein, in T Cell Development, Maturation, and Homeostasis

Zheng Xing; Edward M. Conway; Chulho Kang; Astar Winoto

Survivin is an inhibitor of apoptosis protein that also functions during mitosis. It is expressed in all common tumors and tissues with proliferating cells, including thymus. To examine its role in apoptosis and proliferation, we generated two T cell–specific survivin-deficient mouse lines with deletion occurring at different developmental stages. Analysis of early deleting survivin mice showed arrest at the pre–T cell receptor proliferating checkpoint. Loss of survivin at a later stage resulted in normal thymic development, but peripheral T cells were immature and significantly reduced in number. In contrast to in vitro studies, loss of survivin does not lead to increased apoptosis. However, newborn thymocyte homeostatic and mitogen-induced proliferation of survivin-deficient T cells were greatly impaired. These data suggest that survivin is not essential for T cell apoptosis but is crucial for T cell maturation and proliferation, and survivin-mediated homeostatic expansion is an important physiological process of T cell development.


Diabetes | 2009

Adipose Overexpression of Desnutrin Promotes Fatty Acid Use and Attenuates Diet-Induced Obesity

Maryam Ahmadian; Robin E. Duncan; Krista A. Varady; Danúbia Frasson; Marc K. Hellerstein; Andreas L. Birkenfeld; Varman T. Samuel; Gerald I. Shulman; Yuhui Wang; Chulho Kang; Hei Sook Sul

OBJECTIVE To investigate the role of desnutrin in adipose tissue triacylglycerol (TAG) and fatty acid metabolism. RESEARCH DESIGN AND METHODS We generated transgenic mice overexpressing desnutrin (also called adipose triglyceride lipase [ATGL]) in adipocytes (aP2-desnutrin) and also performed adenoviral-mediated overexpression of desnutrin in 3T3-L1CARΔ1 adipocytes. RESULTS aP2-desnutrin mice were leaner with decreased adipose tissue TAG content and smaller adipocyte size. Overexpression of desnutrin increased lipolysis but did not result in increased serum nonesterified fatty acid levels or ectopic TAG storage. We found increased cycling between diacylglycerol (DAG) and TAG and increased fatty acid oxidation in adipocytes from these mice, as well as improved insulin sensitivity. CONCLUSIONS We show that by increasing lipolysis, desnutrin overexpression causes reduced adipocyte TAG content and attenuation of diet-induced obesity. Desnutrin-mediated lipolysis promotes fatty acid oxidation and re-esterification within adipocytes.


The EMBO Journal | 2003

Transcriptional activation of known and novel apoptotic pathways by Nur77 orphan steroid receptor.

Arvind Rajpal; Yuri A. Cho; Biana Yelent; Petra Koza-Taylor; Dongling Li; Elaine Chen; Michael Whang; Chulho Kang; Thomas G. Turi; Astar Winoto

Nur77 is a nuclear orphan steroid receptor that has been implicated in negative selection. Expression of Nur77 in thymocytes and cell lines leads to apoptosis through a mechanism that remains unclear. In some cell lines, Nur77 was reported to act through a transcription‐independent mechanism involving translocation to mitochondria, leading to cytochrome c release. However, we show here that Nur77‐mediated apoptosis in thymocytes does not involve cytoplasmic cytochrome c release and cannot be rescued by Bcl‐2. Microarray analysis shows that Nur77 induces many genes, including two novel genes (NDG1, NDG2) and known apoptotic genes FasL and TRAIL. Characterization of NDG1 and NDG2 indicates that NDG1 initiates a novel apoptotic pathway in a Bcl‐2‐independent manner. Thus Nur77‐mediated apoptosis in T cells involves Bcl‐2 independent transcriptional activation of several known and novel apoptotic pathways.


Proceedings of the National Academy of Sciences of the United States of America | 2001

T cell-specific FADD-deficient mice: FADD is required for early T cell development

Nisha H. Kabra; Chulho Kang; Lianne C. Hsing; Jianke Zhang; Astar Winoto

FADD/Mort1, initially identified as a Fas-associated death-domain containing protein, functions as an adapter molecule in apoptosis initiated by Fas, tumor necrosis factor receptor-I, DR3, and TRAIL-receptors. However, FADD likely participates in additional signaling cascades. FADD-null mutations in mice are embryonic-lethal, and analysis of FADD−/− T cells from RAG-1−/− reconstituted chimeras has suggested a role for FADD in proliferation of mature T cells. Here, we report the generation of T cell-specific FADD-deficient mice via a conditional genomic rescue approach. We find that FADD-deficiency leads to inhibition of T cell development at the CD4−CD8− stage and a reduction in the number of mature T cells. The FADD mutation does not affect apoptosis or the proximal signaling events of the pre-T cell receptor; introduction of a T cell receptor transgene fails to rescue the mutant phenotype. These data suggest that FADD, through either a death-domain containing receptor or a novel receptor-independent mechanism, is required for the proliferative phase of early T cell development.


Molecular and Cellular Biology | 2003

Occupancy and Function of the 150 Sterol Regulatory Element and 65 E-Box in Nutritional Regulation of the Fatty Acid Synthase Gene in Living Animals

María Jesús Latasa; Michael J. Griffin; Yang Soo Moon; Chulho Kang; Hei Sook Sul

ABSTRACT Upstream regulatory factor (USF) and sterol regulatory element binding protein (SREBP) play key roles in the transcriptional regulation of the fatty acid synthase (FAS) gene by feeding and insulin. Due to the dual binding specificity of SREBP, as well as the presence of multiple consensus sites for these transcription factors in the FAS promoter, their physiologically relevant functional binding sites have been controversial. Here, in order to determine the occupancy of the putative USF and SREBP binding sites, we examined their protein-DNA interactions in living animals by using formaldehyde cross-linking and immunoprecipitation of chromatin and tested the function of these elements by employing mice transgenic for a reporter gene driven by various 5′ deletions as well as site-specific mutations of the FAS promoter. We show that the −332 and −65 E-boxes are bound by USF in both fasted and refed mice, while the −150 SRE is bound by SREBP-1 only in refed mice. We also found that mutation of either the −150 SRE or the −65 E-box abolishes the feeding-induced activation of the FAS promoter in transgenic mice. Furthermore, in vivo occupancy of the FAS promoter by SREBP in the fed state can be prevented by mutation not only of the −150 SRE but, unexpectedly, of the −65 E-box as well. We conclude that the FAS promoter is activated during refeeding via the induced binding of SREBP to the −150 SRE and that USF binding to the −65 E-box is also required for SREBP binding and activation of the FAS promoter.


Science | 2008

Recognition of a Ubiquitous Self Antigen by Prostate Cancer-Infiltrating CD8+ T Lymphocytes

Peter A. Savage; Keith Vosseller; Chulho Kang; Kevin Larimore; Elyn Riedel; Kathleen Wojnoonski; Achim A. Jungbluth; James P. Allison

Substantial evidence exists that many tumors can be specifically recognized by CD8+ T lymphocytes. The definition of antigens targeted by these cells is paramount for the development of effective immunotherapeutic strategies for treating human cancers. In a screen for endogenous tumor-associated T cell responses in a primary mouse model of prostatic adenocarcinoma, we identified a naturally arising CD8+ T cell response that is reactive to a peptide derived from histone H4. Despite the ubiquitous nature of histones, T cell recognition of histone H4 peptide was specifically associated with the presence of prostate cancer in these mice. Thus, the repertoire of antigens recognized by tumor-infiltrating T cells is broader than previously thought and includes peptides derived from ubiquitous self antigens that are normally sequestered from immune detection.


Journal of Experimental Medicine | 2004

Pax5 Activates Immunoglobulin Heavy Chain V to DJ Rearrangement in Transgenic Thymocytes

Lih Yun Hsu; Hong Erh Liang; Kristen Johnson; Chulho Kang; Mark S. Schlissel

Mice deficient for the B cell–restricted transcription factor Pax5 show a defect in the VH to DJH rearrangement step of immunoglobulin heavy chain gene assembly even though the expression of the V(D)J recombinase is not diminished in Pax5 −/− pro–B cells. To investigate whether Pax5 is limiting for VH to DJH rearrangement, we generated transgenic mice which express Pax5 in developing thymocytes. We show that enforced expression of Pax5 in thymocytes results in a partial block in T cell development due to defective pre-TCR signaling in β-selection. Moreover, our results demonstrate that expression of Pax5 in early thymocytes is sufficient to induce VH to DJH rearrangements in CD4+CD8+ T cells and lead us to suggest that Pax5 may play a direct role in the lineage-specific regulation of immunoglobulin heavy chain gene rearrangement.

Collaboration


Dive into the Chulho Kang's collaboration.

Top Co-Authors

Avatar

Hei Sook Sul

University of California

View shared research outputs
Top Co-Authors

Avatar

Astar Winoto

University of California

View shared research outputs
Top Co-Authors

Avatar

Maryam Ahmadian

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Yuhui Wang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kee-Hong Kim

University of California

View shared research outputs
Top Co-Authors

Avatar

Ling Xue

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge