Network


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

Hotspot


Dive into the research topics where Chia-Lin Chen is active.

Publication


Featured researches published by Chia-Lin Chen.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Pluripotency factor-mediated expression of the leptin receptor (OB-R) links obesity to oncogenesis through tumor-initiating stem cells

Douglas Feldman; Chia-Lin Chen; Vasu Punj; Hidekazu Tsukamoto; Keigo Machida

Misregulation of a pluripotency-associated transcription factor network in adult tissues is associated with the expansion of rare, highly malignant tumor-initiating stem cells (TISCs) through poorly understood mechanisms. We demonstrate that robust and selective expression of the receptor for the adipocyte-derived peptide hormone leptin (OB-R) is a characteristic feature of TISCs and of a broad array of embryonic and induced pluripotent stem cells and is mediated directly by the core pluripotency-associated transcription factors OCT4 and SOX2. TISCs exhibit sensitized responses to leptin, including the phosphorylation and activation of the pluripotency-associated oncogene STAT3 and induction of Oct4 and Sox2, thereby establishing a self-reinforcing signaling module. Exposure of cultured mouse embryonic stem cells to leptin sustains pluripotency in the absence of leukemia inhibitory factor. By implanting TISCs into leptin-deficient ob/ob mice or into comparably overweight Leprdb/db mice that produce leptin, we provide evidence of a central role for the leptin-TISC–signaling axis in promoting obesity-induced tumor growth. Differential responses to extrinsic, adipocyte-derived cues may promote the expansion of tumor cell subpopulations and contribute to oncogenesis.


Cell Metabolism | 2016

NANOG Metabolically Reprograms Tumor-Initiating Stem-like Cells through Tumorigenic Changes in Oxidative Phosphorylation and Fatty Acid Metabolism

Chia-Lin Chen; Dinesh Babu Uthaya Kumar; Vasu Punj; Jun Xu; Linda Sher; Stanley M. Tahara; Sonja Hess; Keigo Machida

Stem cell markers, including NANOG, have been implicated in various cancers; however, the functional contribution of NANOG to cancer pathogenesis has remained unclear. Here, we show that NANOG is induced by Toll-like receptor 4 (TLR4) signaling via phosphorylation of E2F1 and that downregulation of Nanog slows down hepatocellular carcinoma (HCC) progression induced by alcohol western diet and hepatitis C virus protein in mice. NANOG ChIP-seq analyses reveal that NANOG regulates the expression of genes involved in mitochondrial metabolic pathways required to maintain tumor-initiating stem-like cells (TICs). NANOG represses mitochondrial oxidative phosphorylation (OXPHOS) genes, as well as ROS generation, and activates fatty acid oxidation (FAO) to support TIC self-renewal and drug resistance. Restoration of OXPHOS activity and inhibition of FAO renders TICs susceptible to a standard care chemotherapy drug for HCC, sorafenib. This study provides insights into the mechanisms of NANOG-mediated generation of TICs, tumorigenesis, and chemoresistance through reprogramming of mitochondrial metabolism.


Hepatology | 2015

Osteopontin deficiency does not prevent but promotes alcoholic neutrophilic hepatitis in mice

Raul Lazaro; Raymond P. Wu; Sun Young Lee; Nian-Ling Zhu; Chia-Lin Chen; Samuel W. French; Jun Xu; Keigo Machida; Hidekazu Tsukamoto

Alcoholic hepatitis (AH) is a distinct spectrum of alcoholic liver disease (ALD) with intense neutrophilic (polymorphonuclear; PMN) inflammation and high mortality. Although a recent study implicates osteopontin (SPP1) in AH, SPP1 is also shown to have protective effects on experimental ALD. To address this unsettled question, we examined the effects of SPP1 deficiency in male mice given 40% calories derived from ad libitum consumption of the Western diet high in cholesterol and saturated fat and the rest from intragastric feeding of alcohol diet without or with weekly alcohol binge. Weekly binge in this new hybrid feeding model shifts chronic ASH with macrophage inflammation and perisinusoidal and pericellular fibrosis to AH in 57% (15 of 26) of mice, accompanied by inductions of chemokines (Spp1, Cxcl1, and interleukin [Il]‐17a), progenitor genes (Cd133, Cd24, Nanog, and epithelial cell adhesion molecule), PMN infiltration, and clinical features of AH, such as hypoalbuminemia, bilirubinemia, and splenomegaly. SPP1 deficiency does not reduce AH incidence and inductions of progenitor and fibrogenic genes, but rather enhances the Il‐17a induction and PMN infiltration in some mice. Furthermore, in the absence of SPP1, chronic ASH mice without weekly binge begin to develop AH. Conclusion: These results suggest that SPP1 has a protective, rather than causal, role for experimental AH reproduced in our model. (Hepatology 2015;61:129–140)


Journal of Gastroenterology and Hepatology | 2012

Cancer stem cells generated by alcohol, diabetes, and hepatitis C virus

Keigo Machida; Chia-Lin Chen; Jian-Chang Liu; Claudine Kashiwabara; Douglas Feldman; Samuel W. French; Linda Sher; Jeong Joseph Hyeongnam; Hidekazu Tsukamoto

Cancer stem cells (tumor‐initiating stem‐like cells: TISCs) are resistant to chemotherapy and are associated with metastatic hepatocellular carcinoma (HCC), which is commonly observed in hepatitis C virus (HCV)‐infected patients with obesity or alcohol abuse. However, it is unknown whether the TLR4‐NANOG pathway serves as a universal oncogenic signaling in the genesis of TISCs and HCC. We aimed to determine whether Tlr4 is a putative proto‐oncogene for TISCs in liver oncogenesis due to different etiologies and how Tlr4 is regulated at the transcriptional and epigenetic levels. CD133+/CD49f+ TISCs were isolated using FACS from HCC developed in HCV Core Tg mice fed alcohol, diethylnitrosamine‐treated mice, and alcoholic patients with or without HCV infection. CD133+/CD49f+ cells isolated from the animal models and patients are tumorigenic both in vitro and in a xenograft model, and Tlr4 or Nanog silencing with shRNA attenuates their tumor initiating property. Functional oncogene screening of a cDNA library identified the organ size control pathway targets Yap1 and AKT activator Igf2bp3 as NANOG‐dependent genes that inhibit transforming growth factor‐β signaling in TISCs. Tlr4 expression is higher in TISCs compared with CD133‐/CD49f+ cells. Taken together, Tlr4 may be a universal proto‐oncogene responsible for the genesis of TLR4‐NANOG dependent TISCs, and this pathway serves as a novel therapeutic target for HCC.


Gastroenterology | 2016

TLR4 Signaling via NANOG Cooperates With STAT3 to Activate Twist1 and Promote Formation of Tumor-initiating Stem-like Cells in Livers of Mice.

Dinesh Babu Uthaya Kumar; Chia-Lin Chen; Jian-Chang Liu; Douglas Feldman; Linda Sher; Samuel W. French; Joseph DiNorcia; Bita V. Naini; Sunhawit Junrungsee; Vatche G. Agopian; Ali Zarrinpar; Keigo Machida

BACKGROUND & AIMS Obesity and alcohol consumption contribute to steatohepatitis, which increases the risk for hepatitis C virus (HCV)-associated hepatocellular carcinomas (HCCs). Mouse hepatocytes that express HCV-NS5A in liver up-regulate the expression of Toll-like receptor 4 (TLR4), and develop liver tumors containing tumor-initiating stem-like cells (TICs) that express NANOG. We investigated whether the TLR4 signals to NANOG to promote the development of TICs and tumorigenesis in mice placed on a Western diet high in cholesterol and saturated fat (HCFD). METHODS We expressed HCV-NS5A from a transgene (NS5A Tg) in Tlr4-/- (C57Bl6/10ScN), and wild-type control mice. Mice were fed a HCFD for 12 months. TICs were identified and isolated based on being CD133+, CD49f+, and CD45-. We obtained 142 paraffin-embedded sections of different stage HCCs and adjacent nontumor areas from the same patients, and performed gene expression, immunofluorescence, and immunohistochemical analyses. RESULTS A higher proportion of NS5A Tg mice developed liver tumors (39%) than mice that did not express HCV NS5A after the HCFD (6%); only 9% of Tlr4-/- NS5A Tg mice fed HCFD developed liver tumors. Livers from NS5A Tg mice fed the HCFD had increased levels of TLR4, NANOG, phosphorylated signal transducer and activator of transcription (pSTAT3), and TWIST1 proteins, and increases in Tlr4, Nanog, Stat3, and Twist1 messenger RNAs. In TICs from NS5A Tg mice, NANOG and pSTAT3 directly interact to activate expression of Twist1. Levels of TLR4, NANOG, pSTAT3, and TWIST were increased in HCC compared with nontumor tissues from patients. CONCLUSIONS HCFD and HCV-NS5A together stimulated TLR4-NANOG and the leptin receptor (OB-R)-pSTAT3 signaling pathways, resulting in liver tumorigenesis through an exaggerated mesenchymal phenotype with prominent Twist1-expressing TICs.


Nature Communications | 2017

Hepatitis C virus has a genetically determined lymphotropism through co-receptor B7.2

Chia-Lin Chen; Jeffrey Y. Huang; Chun-Hsiang Wang; Stanley M. Tahara; Lin Zhou; Yasuteru Kondo; Joel E. Schechter; Lishan Su; Michael M. C. Lai; Takaji Wakita; François-Loïc Cosset; Jae U. Jung; Keigo Machida

B-cell infection by hepatitis C virus (HCV) has been a controversial topic. To examine whether HCV has a genetically determined lymphotropism through a co-receptor specific for the infection by lymphotropic HCV, we established an infectious clone and chimeric virus of hepatotropic and lymphotropic HCV strains derived from an HCV-positive B-cell lymphoma. The viral envelope and 5′-UTR sequences of the lymphotropic HCV strain were responsible for the lymphotropism. Silencing of the virus sensor, RIGI, or overexpression of microRNA-122 promoted persistent viral replication in B cells. By cDNA library screening, we identified an immune cell-specific, co-stimulatory receptor B7.2 (CD86) as a co-receptor of lymphotropic HCV. Infection of B cells by HCV inhibited the recall reaction to antigen stimulation. Together, a co-receptor B7.2 enabled lymphotropic HCV to infect memory B cells, leading to inhibition of memory B-cell function and persistent HCV infection in HCV-infected hosts.


PLOS ONE | 2013

The TBC1D15 oncoprotein controls stem cell self-renewal through destabilization of the Numb-p53 complex.

Douglas Feldman; Chia-Lin Chen; Vasu Punj; Keigo Machida

Stem cell populations are maintained through self-renewing divisions in which one daughter cell commits to a specific fate while the other retains the multipotent characteristics of its parent. The p53 tumor suppressor, in conjunction with its interacting partner protein Numb, preserves this asymmetry and functions as a vital barrier against the unchecked expansion of tumor stem cell pools; however, little is known about the biological control of the Numb-p53 interaction. We show here that Numb and p53 are the constituents of a high molecular mass complex, which is disintegrated upon activation of aPKCζ, a Numb kinase. Using large-scale affinity purification and tandem mass spectrometry, we identify TBC1D15 as a Numb-associated protein and demonstrate that its amino-terminal domain disengages p53 from Numb, triggering p53 proteolysis and promoting self-renewal and pluripotency. Cellular levels of TBC1D15 are diminished upon acute nutrient deprivation through autophagy-mediated degradation, indicating that TBC1D15 serves as a conduit through which cellular metabolic status is linked to self-renewal. The profound deregulation of TBC1D15 expression exhibited in a diverse array of patient tumors underscores its proposed function as an oncoprotein.


Journal of Gastroenterology and Hepatology | 2012

Cancer stem cells generated by alcohol, diabetes, and HCV

Keigo Machida; Chia-Lin Chen; Jian-Chang Liu; Claudine Kashiwabara; Douglas Feldman; Samuel W. French; Linda Sher; Jeong Joseph Hyeongnam; Hidekazu Tsukamoto

Cancer stem cells (tumor‐initiating stem‐like cells: TISCs) are resistant to chemotherapy and are associated with metastatic hepatocellular carcinoma (HCC), which is commonly observed in hepatitis C virus (HCV)‐infected patients with obesity or alcohol abuse. However, it is unknown whether the TLR4‐NANOG pathway serves as a universal oncogenic signaling in the genesis of TISCs and HCC. We aimed to determine whether Tlr4 is a putative proto‐oncogene for TISCs in liver oncogenesis due to different etiologies and how Tlr4 is regulated at the transcriptional and epigenetic levels. CD133+/CD49f+ TISCs were isolated using FACS from HCC developed in HCV Core Tg mice fed alcohol, diethylnitrosamine‐treated mice, and alcoholic patients with or without HCV infection. CD133+/CD49f+ cells isolated from the animal models and patients are tumorigenic both in vitro and in a xenograft model, and Tlr4 or Nanog silencing with shRNA attenuates their tumor initiating property. Functional oncogene screening of a cDNA library identified the organ size control pathway targets Yap1 and AKT activator Igf2bp3 as NANOG‐dependent genes that inhibit transforming growth factor‐β signaling in TISCs. Tlr4 expression is higher in TISCs compared with CD133‐/CD49f+ cells. Taken together, Tlr4 may be a universal proto‐oncogene responsible for the genesis of TLR4‐NANOG dependent TISCs, and this pathway serves as a novel therapeutic target for HCC.


Cancer Research | 2016

Abstract 4657: Selective inhibitors epigenetically modify and eradicate tumor-initiating stem-like cells through downregulating microRNA 22-mediated TET induction and apoptosis

Chia-Lin Chen; Suresh Kumar Swaminathan; Ameya R. Kirtane; Jayanth Panyam; Keigo Machida

Tumor-initiating stem-like cells (TICs) are a minor population in bulk tumors that play a critical role in tumor recurrence and therapy-resistance. We previously showed that a key stemness marker Nanog is upregulated that regulates self-renewal and pluripotency of embryonic stem cells and TICs. We demonstrated that alcohol-mediated activation of Toll-like receptor 4 (TLR4) by endotoxin resulted in increased expression of pluripotency stem cell transcription factor NANOG that metabolically reprograms tumor-initiating stem-like cells (TICs: cancer stem cells) via inhibition of oxidative phosphorylation and activation of fatty acid oxidation in our recently accepted Cell Metabolism paper. Discovery of a drug that specifically targets TICs would be a vital goal for cancer therapy. To identify selective TIC inhibitors, we conducted a high throughput screening of an FDA-approved drug library using combination of Nanog promoter-GFP-based screening with viability-based screening and their combination screening of each hit compound. Our high-throughput screening identified the best combination of repurposed FDA-approved drugs: all-trans retinoic acid (ATRA) and the histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) that repressed NANOG and induces apoptosis of TICs. Treatment of ATRA inhibits self-renewal ability in vitro. To specifically target the CD133 (+) population, we encapsulated ATRA into nanoparticles conjugated with CD133 antibody using biodegradable poly(D,L-lactide-co-glycolide) (PLGA) polymer. The combination treatment significantly inhibited tumor growth compared to single drug treatments while the ATRA or SAHA monotherapy groups did not reduce or even promoted tumor growth. The combination of ATRA with SAHA specifically induces apoptosis of TICs. Targeting of TICs by NANOG antagonism increases drug susceptibility in tumor-bearing mice and achieves ∼90% tumor growth suppression. RNA-seq analysis identified that ATRA+SAHA treatment reduced expression of MicroRNA-22 (miR-22), leading to induction of PTEN and ten-eleven translocation enzymes (TET2), which demethylates DNA. PTEN induction promoted P-FOXO3A and induced BIM, leading to induction of apoptosis. The combined treatment Induced TET1/2 to demethylate p53-binding sites of NANOG promoter, leading to downregulation of NANOG. Taken together, combination treatment of ATRA with SAHA may serve as a novel avenue for HCC treatment. Novel combination of repurposed drugs is cost-effective therapeutic strategy to target microRNA for eradication of TICs, leading to inhibition of metastasis and recurrence. Citation Format: Chia-Lin Chen, Suresh Swaminathan, Ameya Kirtane, Jayanth Panyam, Keigo Machida. Selective inhibitors epigenetically modify and eradicate tumor-initiating stem-like cells through downregulating microRNA 22-mediated TET induction and apoptosis. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4657.


Cancer Research | 2015

Abstract 3040: NANOG metabolically reprograms tumor-initiating stem-like cells in oxidative phosphorylation and fatty acid metabolism

Chia-Lin Chen; Dinesh Babu Uthaya Kumar; Vasu Punj; Jun Xu; Linda Sher; Sonja Hess; Keigo Machida

Stem cell markers such as NANOG have been implicated in various cancers; however, the functional contribution of pluripotency transcription factor NANOG to cancer pathogenesis has remained unclear. Gene profiling, proteomics and metabolomics approaches were combined to identify the pathway(s) altered in tumors. Here, we show that Toll-like receptor 4 (TLR4) signaling phosphorylates E2F1 to transactivate NANOG via TAK1/TBK1. CD133+/CD49f+ TICs isolated from human HCC have tumorigenic activities dependent on TLR4 and NANOG. Down-regulation of Nanog significantly reduces tumor development in obesity-associated HCC mouse model. In the search for the NANOG-dependent mechanisms underlying growth of tumor-initiating stem-like cells (TICs), NANOG ChIP-seq identifies the genes associated with mitochondrial metabolic pathways. The specific pathways, which were examined: oxidative phosphorylation (OXPHOS) and fatty acid oxidation were identified as novel NANOG-mediated oncogenic pathways by NANOG ChIP-seq analysis and metabolomics. The causal roles of NANOG in mitochondrial metabolic reprogramming occurs through the inhibition of oxidative phosphorylation (OXPHOS) and levels of mitochondrial ROS and activation of fatty acid oxidation (FAO), which are required for both the self-renewal of TICs and their drug resistance. PPARδ physically interacts with NANOG. Restoration of OXPHOS and inhibition of FAO rendered TICs susceptible to drug. Our data showed that NANOG reprogramming of mitochondrial metabolism was responsible for human TIC oncogenicity and chemo-resistance. Together, NANOG-mediated metabolic reprogramming through dysregulation of mitochondrial functions generates TICs and drives tumorigenesis, providing insight into mechanisms of NANOG-mediated metabolic reprogramming and its roles in TIC oncogenic activity and chemo-resistance. NANOG suppresses OXPHOS and activates FAO, thus inhibiting OCR and ROS production, conferring a tumor chemoresistant state. These novel pathways are potential drug targets for the highly malignant TICs found in cancer patients. Reversal of NANOG-dependent effects on OXPHOS and FAO gene may offer a noteworthy strategy of countering therapeutic drug resistance associated with NANOG activation. Citation Format: Chia-Lin Chen, Dinesh Babu Uthaya Kumar, Vasu Punj, Jun Xu, Linda Sher, Sonja Hess, Keigo Machida. NANOG metabolically reprograms tumor-initiating stem-like cells in oxidative phosphorylation and fatty acid metabolism. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3040. doi:10.1158/1538-7445.AM2015-3040

Collaboration


Dive into the Chia-Lin Chen's collaboration.

Top Co-Authors

Avatar

Keigo Machida

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Linda Sher

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Hidekazu Tsukamoto

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Douglas Feldman

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Jian-Chang Liu

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Vasu Punj

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claudine Kashiwabara

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Dinesh Babu Uthaya Kumar

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Jeong Joseph Hyeongnam

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge