1
|
He X, Liu Y, Gao X, Tang F, Tian Y, Gong S, Shen J, Wang A, Sun L, Wei W, Weng L. N-terminal acetylation of transcription factor LIP induces immune therapy resistance via suppression of PD-L1 expression in non-small cell lung cancer. J Immunother Cancer 2024; 12:e009905. [PMID: 39615895 PMCID: PMC11624798 DOI: 10.1136/jitc-2024-009905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/05/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND Programmed death-1 (PD-1) checkpoint blockade has revolutionized cancer therapy, yet its clinical success is confined to a subset of patients, underscoring the urgent need to understand the molecular underpinnings of programmed cell death ligand 1 (PD-L1) expression to combat immunotherapy resistance. METHODS Employing CRISPR/Cas9 screening, we identified key regulators of PD-L1 in non-small cell lung cancer (NSCLC) cells, focusing on the transcription factor CEBPB and its isoform liver-enriched inhibitory protein (LIP). Through chromatin immunoprecipitation (ChIP) and luciferase reporter assays, we explored the interaction between LIP and basic-helix-loop-helix E22 (BHLHE22) in controlling PD-L1 transcription. We also used immunofluorescence and NBD-CI assays to examine how N-terminal acetylation affects LIP's subcellular localization. The impact of LIP on tumor growth was assessed via subcutaneous tumorigenicity assays, while immunohistochemistry and immunofluorescence were used to analyze LIP-induced alterations in the tumor immune microenvironment. RESULTS Our research indicates that CEBPB, particularly its LIP isoform, significantly suppresses PD-L1 expression in NSCLC cells. This suppression is contingent on LIP's N-terminal acetylation by the N-terminal acetyltransferase A complex, which facilitates LIP's nuclear entry and interaction with BHLHE22. This interaction leads to the formation of a co-repressor complex at the PD-L1 promoter, effectively reducing PD-L1 expression and enhancing the tumor immune response. CONCLUSIONS Identifying CEBPB, especially the LIP isoform, as a pivotal regulator of PD-L1 expression sheds light on the mechanisms behind PD-1 blockade resistance in NSCLC. Our findings suggest that modulating LIP's function or its molecular interactions might offer a novel approach to boosting the efficacy of immunotherapies.
Collapse
Affiliation(s)
- Xiang He
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Cancer Center, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| | - Yongshuo Liu
- Department of Pathology and Lab Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xing Gao
- Department of Stomatology, Xiangya Hospital Central South University, Changsha, Hunan, China
- Center of Oral and Maxillofacial Cancer (COMAC), Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Feiyu Tang
- Center for Biotherapy, Sun Yat-Sen University, Guangzhou, China
| | - Yuxi Tian
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Siyuan Gong
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Cancer Center, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| | - Jia Shen
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Cancer Center, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| | - Aimin Wang
- Department of Emergency, Xiangya Hospital Central South University, Changsha, China
| | - Lunquan Sun
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Cancer Center, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| | - Wensheng Wei
- Biomedical Pioneering Innovation Center, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, Peking University School of Life Sciences, Beijing, China
- Changping Laboratory, Beijing, China
| | - Liang Weng
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| |
Collapse
|
2
|
Wang CZ, Zhang ZQ, Zhang Y, Zheng LF, Liu Y, Yan AT, Zhang YC, Chang QH, Sha S, Xu ZJ. Comprehensive characterization of TGFB1 across hematological malignancies. Sci Rep 2023; 13:19107. [PMID: 37925591 PMCID: PMC10625629 DOI: 10.1038/s41598-023-46552-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/02/2023] [Indexed: 11/05/2023] Open
Abstract
TGFB1, which encodes TGF-β1, a potent cytokine regulating varies cellular processes including immune responses. TGF-β1 plays context-dependent roles in cancers and is increasingly recognized as a therapeutic target to enhance immunotherapy responses. We comprehensively evaluated expression of TGFB1 and its clinical and biological effects across hematological malignancies. TGFB1 expression was first explored using data from the GTEx, CCLE, and TCGA databases. The expression and clinical significances of TGFB1 in hematological malignancies were analyzed using Hemap and our In Silico curated datasets. We also analyzed the relationship between TGFB1 with immune scores and immune cell infiltrations in Hemap. We further assessed the value of TGFB1 in predicting immunotherapy response using TIDE and real-world immunotherapy datasets. TGFB1 showed a hematologic-tissue-specific expression pattern both across normal tissues and cancer types. TGFB1 expression were broadly dysregulated in blood cancers and generally associated with adverse prognosis. TGFB1 expression were associated with distinct TME properties among different blood cancer types. In addition, TGFB1 expression was found to be a useful marker in predicting immunotherapy responses. Our results suggest that TGFB1 is broadly dysregulated in hematological malignancies. TGFB1 might regulate the immune microenvironment in a cancer-type-specific manner, which could be applied in the development of new targeted drugs for immunotherapy.
Collapse
Affiliation(s)
- Cui-Zhu Wang
- Department of Hematology and Oncology, Affiliated Haian Hospital of Nantong University, Nantong, China
| | - Zi-Qi Zhang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yan Zhang
- Department of Hematology and Oncology, Affiliated Haian Hospital of Nantong University, Nantong, China
| | - Liang-Feng Zheng
- Laboratory Center, Affiliated Haian Hospital of Nantong University, Nantong, China
| | - Yang Liu
- Clinical Nutrition Department, Haian Hospital of Traditional Chinese Medicine, Nantong, China
| | - Ai-Ting Yan
- Department of Hematology and Oncology, Affiliated Haian Hospital of Nantong University, Nantong, China
| | - Yuan-Cui Zhang
- Department of Respiratory Medicine, The Affiliated Zhenjiang Third Hospital of Jiangsu University, 1 Dingmao Bridge, Youth Square, Zhenjiang, 212002, China
| | - Qing-Hua Chang
- Department of Respiratory Medicine, The Affiliated Zhenjiang Third Hospital of Jiangsu University, 1 Dingmao Bridge, Youth Square, Zhenjiang, 212002, China.
| | - Suo Sha
- Surgery of Traditional Chinese Medicine, Haian Hospital of Traditional Chinese Medicine, Nantong, 226600, China.
| | - Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang, 212002, China.
| |
Collapse
|
3
|
Ding Z, Shi R, Hu W, Tian L, Sun R, Wu Y, Zhang X. Cancer-associated fibroblasts in hematologic malignancies: elucidating roles and spotlighting therapeutic targets. Front Oncol 2023; 13:1193978. [PMID: 37746306 PMCID: PMC10511871 DOI: 10.3389/fonc.2023.1193978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Hematologic malignancies comprise a diverse range of blood, bone marrow, and organ-related disorders that present significant challenges due to drug resistance, relapse, and treatment failure. Cancer-associated fibroblasts (CAFs) represent a critical component of the tumor microenvironment (TME) and have recently emerged as potential therapeutic targets. In this comprehensive review, we summarize the latest findings on the roles of CAFs in various hematologic malignancies, including acute leukemia, multiple myeloma, chronic lymphocytic leukemia, myeloproliferative neoplasms, and lymphoma. We also explore their involvement in tumor progression, drug resistance, and the various signaling pathways implicated in their activation and function. While the underlying mechanisms and the existence of multiple CAF subtypes pose challenges, targeting CAFs and their associated pathways offers a promising avenue for the development of innovative treatments to improve patient outcomes in hematologic malignancies.
Collapse
Affiliation(s)
- Ziyang Ding
- The Second Clinical School of Nanjing Medical University, Nanjing, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weikang Hu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Tian
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rong Sun
- Department of Radiation Oncology, Jinling Hospital, Nanjing, China
| | - Yang Wu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoyan Zhang
- Department of Hematology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Liao Y, Fu Z, Huang Y, Wu S, Wang Z, Ye S, Zeng W, Zeng G, Li D, Yang Y, Pei K, Yang J, Hu Z, Liang X, Hu J, Liu M, Jin J, Cai C. Interleukin-18-primed human umbilical cord-mesenchymal stem cells achieve superior therapeutic efficacy for severe viral pneumonia via enhancing T-cell immunosuppression. Cell Death Dis 2023; 14:66. [PMID: 36707501 PMCID: PMC9883134 DOI: 10.1038/s41419-023-05597-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/29/2023]
Abstract
Coronavirus disease 2019 (COVID-19) treatments are still urgently needed for critically and severely ill patients. Human umbilical cord-mesenchymal stem cells (hUC-MSCs) infusion has therapeutic benefits in COVID-19 patients; however, uncertain therapeutic efficacy has been reported in severe patients. In this study, we selected an appropriate cytokine, IL-18, based on the special cytokine expression profile in severe pneumonia of mice induced by H1N1virus to prime hUC-MSCs in vitro and improve the therapeutic effect of hUC-MSCs in vivo. In vitro, we demonstrated that IL-18-primed hUC-MSCs (IL18-hUCMSC) have higher proliferative ability than non-primed hUC-MSCs (hUCMSCcon). In addition, VCAM-1, MMP-1, TGF-β1, and some chemokines (CCL2 and CXCL12 cytokines) are more highly expressed in IL18-hUCMSCs. We found that IL18-hUCMSC significantly enhanced the immunosuppressive effect on CD3+ T-cells. In vivo, we demonstrated that IL18-hUCMSC infusion could reduce the body weight loss caused by a viral infection and significantly improve the survival rate. Of note, IL18-hUCMSC can also significantly attenuate certain clinical symptoms, including reduced activity, ruffled fur, hunched backs, and lung injuries. Pathologically, IL18-hUCMSC transplantation significantly enhanced the inhibition of inflammation, viral load, fibrosis, and cell apoptosis in acute lung injuries. Notably, IL18-hUCMSC treatment has a superior inhibitory effect on T-cell exudation and proinflammatory cytokine secretion in bronchoalveolar lavage fluid (BALF). Altogether, IL-18 is a promising cytokine that can prime hUC-MSCs to improve the efficacy of precision therapy against viral-induced pneumonia, such as COVID-19.
Collapse
Affiliation(s)
- Yan Liao
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Zeqin Fu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Yinfu Huang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Shiduo Wu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Zhen Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Shaotang Ye
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Weijie Zeng
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Guifang Zeng
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Duanduan Li
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Yulin Yang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Ke Pei
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Jian Yang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Zhiwei Hu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Xiao Liang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Junyuan Hu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China.
| | - Muyun Liu
- National-Local Associated Engineering Laboratory for Personalized Cell Therapy, Shenzhen, 518054, China.
| | - Juan Jin
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310000, China.
| | - Cheguo Cai
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
5
|
Inhibitory Effect of the LY2109761 on the Development of Human Keloid Fibroblasts. ACTA ACUST UNITED AC 2021; 2021:8883427. [PMID: 33628711 PMCID: PMC7889383 DOI: 10.1155/2021/8883427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 12/15/2020] [Accepted: 01/23/2021] [Indexed: 11/18/2022]
Abstract
Keloids are scars characterized by abnormal proliferation of fibroblasts and overproduction of extracellular matrix components including collagen. We previously showed that LY2109761, a transforming growth factor- (TGF-) β receptor inhibitor, suppressed the secretion of matrix components and slowed the proliferation of fibroblasts derived from human hypertrophic scar tissue. However, the exact mechanism underlying this effect remains unclear. Here, we replicated the above results in keloid-derived fibroblasts and show that LY2109761 promoted apoptosis, decreased the phosphorylation of Smad2 and Smad3, and suppressed TGF-β1. These results suggest that the development and pathogenesis of keloids are positively regulated by the Smad2/3 signaling pathway and the upregulation of TGF-β1 receptors. LY2109761 and other inhibitors of these processes may therefore serve as therapeutic targets to limit excessive scarring after injury.
Collapse
|
6
|
Yuan B, El Dana F, Ly S, Yan Y, Ruvolo V, Shpall EJ, Konopleva M, Andreeff M, Battula VL. Bone marrow stromal cells induce an ALDH+ stem cell-like phenotype and enhance therapy resistance in AML through a TGF-β-p38-ALDH2 pathway. PLoS One 2020; 15:e0242809. [PMID: 33253299 PMCID: PMC7703975 DOI: 10.1371/journal.pone.0242809] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
The bone marrow microenvironment (BME) in acute myeloid leukemia (AML) consists of various cell types that support the growth of AML cells and protect them from chemotherapy. Mesenchymal stromal cells (MSCs) in the BME have been shown to contribute immensely to leukemogenesis and chemotherapy resistance in AML cells. However, the mechanism of stroma-induced chemotherapy resistance is not known. Here, we hypothesized that stromal cells promote a stem-like phenotype in AML cells, thereby inducing tumorigenecity and therapy resistance. To test our hypothesis, we co-cultured AML cell lines and patient samples with BM-derived MSCs and determined aldehyde dehydrogenase (ALDH) activity and performed gene expression profiling by RNA sequencing. We found that the percentage of ALDH+ cells increased dramatically when AML cells were co-cultured with MSCs. However, among the 19 ALDH isoforms, ALDH2 and ALDH1L2 were the only two that were significantly upregulated in AML cells co-cultured with stromal cells compared to cells cultured alone. Mechanistic studies revealed that the transforming growth factor-β1 (TGF-β1)-regulated gene signature is activated in AML cells co-cultured with MSCs. Knockdown of TGF-β1 in BM-MSCs inhibited stroma-induced ALDH activity and ALDH2 expression in AML cells, whereas treatment with recombinant TGF-β1 induced the ALDH+ phenotype in AML cells. We also found that TGF-β1-induced ALDH2 expression in AML cells is mediated by the non-canonical pathway through the activation of p38. Interestingly, inhibition of ALDH2 with diadzin and CVT-10216 significantly inhibited MSC-induced ALDH activity in AML cells and sensitized them to chemotherapy, even in the presence of MSCs. Collectively, BM stroma induces ALDH2 activity in AML cells through the non-canonical TGF-β pathway. Inhibition of ALDH2 sensitizes AML cells to chemotherapy.
Collapse
Affiliation(s)
- Bin Yuan
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Fouad El Dana
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Stanley Ly
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Yuanqing Yan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Vivian Ruvolo
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Elizabeth J. Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Marina Konopleva
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Michael Andreeff
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Venkata Lokesh Battula
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
7
|
Çetin İ, Topçul MR. Evaluation of the cytotoxic effect of Ly2109761 on HeLa cells using the xCELLigence RTCA system. Oncol Lett 2019; 17:683-687. [PMID: 30655817 DOI: 10.3892/ol.2018.9556] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 08/15/2018] [Indexed: 02/04/2023] Open
Abstract
In the present study, the in vitro cytotoxic effect of a novel transforming growth factor-β receptor inhibitor, LY2109761, was investigated in the human cervix carcinoma HeLa cell lines. For the purpose of the present study, cell index values obtained using the xCELLigence Real-Time Cell Analysis DP instrument, and mitotic, labelling and apoptotic index analysis were used. The results of the present study indicated that LY2109761 affected the cytoskeleton of HeLa cells, decreased the mitotic and labelling index values of the HeLa cell line, and increased the apoptotic index values. Significant differences were observed between the control group which was not treated with LY2109761 and the experimental groups, which were treated with LY2109761 (P<0.01). The results of the present study suggest that LY2109761 may serve as a promising treatment option for cervix carcinoma.
Collapse
Affiliation(s)
- İdil Çetin
- Department of Biology, Faculty of Science, Istanbul University, Istanbul 34459, Turkey
| | - Mehmet R Topçul
- Department of Biology, Faculty of Science, Istanbul University, Istanbul 34459, Turkey
| |
Collapse
|
8
|
Wei G, Xu Q, Liu L, Zhang H, Tan X, Zhang C, Han C, Guo Y, Han G, Zhang C. LY2109761 reduces TGF-β1-induced collagen production and contraction in hypertrophic scar fibroblasts. Arch Dermatol Res 2018; 310:615-623. [PMID: 30046895 DOI: 10.1007/s00403-018-1849-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/13/2018] [Accepted: 07/17/2018] [Indexed: 10/28/2022]
Abstract
Hypertrophic scars (HS) are fibro-hyperproliferative dermal lesions with effusive continuous accumulation of extracellular matrix components, particularly collagen. They usually occur after dermal injury in genetically susceptible individuals and cause both physical and psychological distress for the affected individuals. Transforming growth factor-β1 (TGF-β1) is known to mediate wound healing process by regulating cell differentiation, collagen production and extracellular matrix degradation. The sustained high expression of TGF-β1 is believed to result in the formation of hypertrophic scars. Inhibition of TGF-β1 signaling pathway may represent one of effective strategies for limiting excessive scarring. LY2109761, an orally active TβRI/II kinase dual inhibitor, has been previously reported that it had inhibitory effects on carcinomas and attenuates Radiation-induced pulmonary murine fibrosis. Our results revealed that LY2109761 reduced TGF-β1-induced collagen production and α-smooth muscle actin (α-SMA) expression, and attenuated TGF-β1-induced cell contraction in hypertrophic scar fibroblasts. The data from this study provide evidence supporting the potential use of LY2109761 as a novel treatment for hypertrophic scars.
Collapse
Affiliation(s)
- Guo Wei
- Department of Dermato-venereology, the Second Hospital of Shandong University, 247 Beiyuan Dajie Street, Jinan, 250033, Shandong, China
| | - Qingqing Xu
- Department of Dermato-venereology, the Second Hospital of Shandong University, 247 Beiyuan Dajie Street, Jinan, 250033, Shandong, China
| | - Lin Liu
- Department of Dermato-venereology, the Second Hospital of Shandong University, 247 Beiyuan Dajie Street, Jinan, 250033, Shandong, China
| | - Huanhuan Zhang
- Department of Dermato-venereology, the Second Hospital of Shandong University, 247 Beiyuan Dajie Street, Jinan, 250033, Shandong, China
| | - Xi Tan
- Department of Dermato-venereology, the Second Hospital of Shandong University, 247 Beiyuan Dajie Street, Jinan, 250033, Shandong, China
| | - Chunhong Zhang
- Department of Dermato-venereology, the Second Hospital of Shandong University, 247 Beiyuan Dajie Street, Jinan, 250033, Shandong, China
| | - Changyu Han
- Department of Dermato-venereology, the Second Hospital of Shandong University, 247 Beiyuan Dajie Street, Jinan, 250033, Shandong, China
| | - Yanxia Guo
- Institute of Medical Sciences, the Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| | - Ganwen Han
- Department of Dermato-venereology, the Second Hospital of Shandong University, 247 Beiyuan Dajie Street, Jinan, 250033, Shandong, China.,Department of Dermatology, Peking University International Hospital, Beijing, 102206, China
| | - Chunmin Zhang
- Department of Dermato-venereology, the Second Hospital of Shandong University, 247 Beiyuan Dajie Street, Jinan, 250033, Shandong, China.
| |
Collapse
|
9
|
Ruvolo PP, Ruvolo VR, Burks JK, Qiu Y, Wang RY, Shpall EJ, Mirandola L, Hail N, Zeng Z, McQueen T, Daver N, Post SM, Chiriva-Internati M, Kornblau SM, Andreeff M. Role of MSC-derived galectin 3 in the AML microenvironment. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2018; 1865:959-969. [PMID: 29655803 PMCID: PMC5936474 DOI: 10.1016/j.bbamcr.2018.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 12/16/2022]
Abstract
In acute myeloid leukemia (AML), high Galectin 3 (LGALS3) expression is associated with poor prognosis. The role of LGALS3 derived from mesenchymal stromal cells (MSC) in the AML microenvironment is unclear; however, we have recently found high LGALS3 expression in MSC derived from AML patients is associated with relapse. In this study, we used reverse phase protein analysis (RPPA) to correlate LGALS3 expression in AML MSC with 119 other proteins including variants of these proteins such as phosphorylated forms or cleaved forms to identify biologically relevant pathways. RPPA revealed that LGALS3 protein was positively correlated with expression of thirteen proteins including MYC, phosphorylated beta-Catenin (p-CTNNB1), and AKT2 and negatively correlated with expression of six proteins including integrin beta 3 (ITGB3). String analysis revealed that proteins positively correlated with LGALS3 showed strong interconnectivity. Consistent with the RPPA results, LGALS3 suppression by shRNA in MSC resulted in decreased MYC and AKT expression while ITGB3 was induced. In co-culture, the ability of AML cell to adhere to MSC LGALS3 shRNA transductants was reduced compared to AML cell adhesion to MSC control shRNA transductants. Finally, use of novel specific LGALS3 inhibitor CBP.001 in co-culture of AML cells with MSC reduced viable leukemia cell populations with induced apoptosis and augmented the chemotherapeutic effect of AraC. In summary, the current study demonstrates that MSC-derived LGALS3 may be critical for important biological pathways for MSC homeostasis and for regulating AML cell localization and survival in the leukemia microenvironmental niche.
Collapse
Affiliation(s)
- Peter P Ruvolo
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Section of Molecular Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| | - Vivian R Ruvolo
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Section of Molecular Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jared K Burks
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Section of Molecular Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - YiHua Qiu
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Section of Molecular Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rui-Yu Wang
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Section of Molecular Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Numsen Hail
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Section of Molecular Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zhihong Zeng
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Section of Molecular Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Teresa McQueen
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Section of Molecular Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Naval Daver
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sean M Post
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maurizio Chiriva-Internati
- Kiromic Biopharma, Houston, TX, United States; Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven M Kornblau
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Section of Molecular Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael Andreeff
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Section of Molecular Hematology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
10
|
de Araújo Farias V, Carrillo-Gálvez AB, Martín F, Anderson P. TGF-β and mesenchymal stromal cells in regenerative medicine, autoimmunity and cancer. Cytokine Growth Factor Rev 2018; 43:25-37. [PMID: 29954665 DOI: 10.1016/j.cytogfr.2018.06.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/12/2018] [Indexed: 12/30/2022]
Abstract
Multipotent mesenchymal stromal cells (MSCs) represent a promising cell-based therapy in regenerative medicine and for the treatment of inflammatory/autoimmune diseases. Importantly, MSCs have emerged as an important contributor to the tumor stroma with both pro- and anti-tumorigenic effects. However, the successful translation of MSCs to the clinic and the prevention of their tumorigenic and metastatic effect require a greater understanding of factors controlling their proliferation, differentiation, migration and immunomodulation in vitro and in vivo. The transforming growth factor(TGF)-β1, 2 and 3 are involved in almost every aspect of MSC function. The aim of this review is to highlight the roles that TGF-β play in the biology and therapeutic applications of MSCs. We will discuss the how TGF-β modulate MSC function as well as the paracrine effects of MSC-derived TGF-β on other cell types in the context of tissue regeneration, immune responses and cancer. Finally, taking all these aspects into consideration we discuss how modulation of TGF-β signaling/production in MSCs could be of clinical interest.
Collapse
Affiliation(s)
- Virgínea de Araújo Farias
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain; Facultad de Odontología, Universidad de Granada, Campus Universitario de Cartuja, 18071 Granada, Spain
| | - Ana Belén Carrillo-Gálvez
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain
| | - Francisco Martín
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain
| | - Per Anderson
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain.
| |
Collapse
|
11
|
Saunders JH, Onion D, Collier P, Dorrington MS, Argent RH, Clarke PA, Reece-Smith AM, Parsons SL, Grabowska AM. Individual patient oesophageal cancer 3D models for tailored treatment. Oncotarget 2018; 8:24224-24236. [PMID: 27736801 PMCID: PMC5421842 DOI: 10.18632/oncotarget.12500] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 09/29/2016] [Indexed: 12/18/2022] Open
Abstract
Background A model to predict chemotherapy response would provide a marked clinical benefit, enabling tailored treatment of oesophageal cancer, where less than half of patients respond to the routinely administered chemotherapy. Methods Cancer cells were established from tumour biopsies taken from individual patients about to undergo neoadjuvant chemotherapy. A 3D-tumour growth assay (3D-TGA) was developed, in which cancer cells were grown with or without supporting mesenchymal cells, then subjected to chemo-sensitivity testing using the standard chemotherapy administered in clinic, and a novel emerging HDAC inhibitor, Panobinostat. RESULTS Individual patients cancer cells could be expanded and screened within a clinically applicable timescale of 3 weeks. Incorporating mesenchymal support within the 3D-TGA significantly enhanced both the growth and drug resistance profiles of the patients cancer cells. The ex vivo drug response in the presence, but not absence, of mesenchymal cells accurately reflected clinical chemo-sensitivity, as measured by tumour regression grade. Combination with Panobinostat enhanced response and proved efficacious in otherwise chemo-resistant tumours. Conclusions This novel method of establishing individual patient oesophageal cancers in the laboratory, from small endoscopic biopsies, enables clinically-relevant chemo-sensitivity testing, and reduces use of animals by providing more refined in vitro models for pre-screening of drugs. The 3D-TGA accurately predicted chemo-sensitivity in patients, and could be developed to guide tailored patient treatment. The incorporation of mesenchymal cells as the stromal cell component of the tumour micro-environment had a significant effect upon enhancing chemotherapy drug resistance in oesophageal cancer, and could prove a useful target for future drug development.
Collapse
Affiliation(s)
- John H Saunders
- Cancer Biology Unit, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK.,Department of Upper GI Surgery, City Hospital Campus, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - David Onion
- Cancer Biology Unit, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Pamela Collier
- Cancer Biology Unit, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Matthew S Dorrington
- Cancer Biology Unit, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Richard H Argent
- Cancer Biology Unit, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Philip A Clarke
- Cancer Biology Unit, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Alex M Reece-Smith
- Cancer Biology Unit, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK.,Department of Upper GI Surgery, City Hospital Campus, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Simon L Parsons
- Cancer Biology Unit, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK.,Department of Upper GI Surgery, City Hospital Campus, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Anna M Grabowska
- Cancer Biology Unit, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
12
|
Schelker RC, Iberl S, Müller G, Hart C, Herr W, Grassinger J. TGF-β1 and CXCL12 modulate proliferation and chemotherapy sensitivity of acute myeloid leukemia cells co-cultured with multipotent mesenchymal stromal cells. Hematology 2017; 23:337-345. [DOI: 10.1080/10245332.2017.1402455] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Roland Christian Schelker
- Department of Internal Medicine III, Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Sabine Iberl
- Department of Internal Medicine III, Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Gunnar Müller
- Department of Internal Medicine III, Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Christina Hart
- Department of Internal Medicine III, Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Jochen Grassinger
- Department of Internal Medicine III, Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
| |
Collapse
|
13
|
Fong G, Backman LJ, Alfredson H, Scott A, Danielson P. The effects of substance P and acetylcholine on human tenocyte proliferation converge mechanistically via TGF-β1. PLoS One 2017; 12:e0174101. [PMID: 28301610 PMCID: PMC5354451 DOI: 10.1371/journal.pone.0174101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/04/2017] [Indexed: 01/10/2023] Open
Abstract
Previous in vitro studies on human tendon cells (tenocytes) have demonstrated that the exogenous administration of substance P (SP) and acetylcholine (ACh) independently result in tenocyte proliferation, which is a prominent feature of tendinosis. Interestingly, the possible link between SP and ACh has not yet been explored in human tenocytes. Recent studies in other cell types demonstrate that both SP and ACh independently upregulate TGF-β1 expression via their respective receptors, the neurokinin 1 receptor (NK-1R) and muscarinic ACh receptors (mAChRs). Furthermore, TGF-β1 has been shown to downregulate NK-1R expression in human keratocytes. The aim of this study was to examine if TGF-β1 is the intermediary player involved in mediating the proliferative pathway shared by SP and ACh in human tenocytes. The results showed that exogenous administration of SP and ACh both caused significant upregulation of TGF-β1 at the mRNA and protein levels. Exposing cells to TGF-β1 resulted in increased cell viability of tenocytes, which was blocked in the presence of the TGFβRI/II kinase inhibitor. In addition, the proliferative effects of SP and ACh on tenocytes were reduced by the TGFβRI/II kinase inhibitor; this supports the hypothesis that the proliferative effects of these signal substances are mediated via the TGF-β axis. Furthermore, exogenous TGF-β1 downregulated NK-1R and mAChRs expression at both the mRNA and protein levels, and these effects were negated by simultaneous exposure to the TGFβRI/II kinase inhibitor, suggesting a negative feedback loop. In conclusion, the results indicate that TGF-β1 is the intermediary player through which the proliferative actions of both SP and ACh converge mechanistically.
Collapse
Affiliation(s)
- Gloria Fong
- Dept. of Integrative Medical Biology, Anatomy, Umeå University, Umeå, Sweden
- Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Ludvig J. Backman
- Dept. of Integrative Medical Biology, Anatomy, Umeå University, Umeå, Sweden
| | - Håkan Alfredson
- Dept. of Community Medicine and Rehabilitation, Sports Medicine, Umeå University, Umeå, Sweden
| | - Alex Scott
- Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Patrik Danielson
- Dept. of Integrative Medical Biology, Anatomy, Umeå University, Umeå, Sweden
- Dept. of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
14
|
Maj M, Bajek A, Nalejska E, Porowinska D, Kloskowski T, Gackowska L, Drewa T. Influence of Mesenchymal Stem Cells Conditioned Media on Proliferation of Urinary Tract Cancer Cell Lines and Their Sensitivity to Ciprofloxacin. J Cell Biochem 2016; 118:1361-1368. [DOI: 10.1002/jcb.25794] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/14/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Malgorzata Maj
- Chair of Urology, Department of Tissue Engineering; Collegium Medicum; Nicolaus Copernicus University; Karlowicza 24 85-092 Bydgoszcz Poland
| | - Anna Bajek
- Chair of Urology, Department of Tissue Engineering; Collegium Medicum; Nicolaus Copernicus University; Karlowicza 24 85-092 Bydgoszcz Poland
| | - Ewelina Nalejska
- Chair of Urology, Department of Tissue Engineering; Collegium Medicum; Nicolaus Copernicus University; Karlowicza 24 85-092 Bydgoszcz Poland
| | - Dorota Porowinska
- Department of Biochemistry; Nicolaus Copernicus University; Gagarina 7 87-100 Torun Poland
| | - Tomasz Kloskowski
- Chair of Urology, Department of Regenerative Medicine; Collegium Medicum, Nicolaus Copernicus University; Sklodowskiej-Curie 9 85-094 Bydgoszcz Poland
| | - Lidia Gackowska
- Department of Immunology; Collegium Medicum; Nicolaus Copernicus University; Sklodowskiej-Curie 9 85-094 Bydgoszcz Poland
| | - Tomasz Drewa
- Chair of Urology, Clinic of General and Oncological Urology; Collegium Medicum, Nicolaus Copernicus University; Sklodowskiej-Curie 9 85-094 Bydgoszcz Poland
- Department of Urology; Nicolaus Copernicus Hospital; Batorego 17/19 87-100 Torun Poland
| |
Collapse
|
15
|
Velaei K, Samadi N, Barazvan B, Soleimani Rad J. Tumor microenvironment-mediated chemoresistance in breast cancer. Breast 2016; 30:92-100. [PMID: 27668856 DOI: 10.1016/j.breast.2016.09.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 09/02/2016] [Accepted: 09/02/2016] [Indexed: 12/20/2022] Open
Abstract
Therapy resistance or tumor relapse in cancer is common. Tumors develop resistance to chemotherapeutic through a variety of mechanisms, with tumor microenvironment (TM) serving pivotal roles. Using breast cancer as a paradigm, we propose that responses of cancer cells to drugs are not exclusively determined by their intrinsic characteristics but are also controlled by deriving signals from TM. Affected microenvironment by chemotherapy is an avenue to promote phenotype which tends to resist on to be ruined. Therefore, exclusively targeting cancer cells does not demolish tumor recurrence after chemotherapy. Regardless of tumor-microenvironment pathways and their profound influence on the responsiveness of treatment, diversity of molecular properties of breast cancer also behave differently in terms of response to chemotherapy. And also it is assumed that there is cross-talk between phenotypic diversity and TM. Collectively, raising complex signal from TM in chemotherapy condition often encourages cancer cells are not killed but strengthen. Here, we summarized how TM modifies responses to chemotherapy in breast cancer. We also discussed successful treatment strategies have been considered TM in breast cancer treatment.
Collapse
Affiliation(s)
- Kobra Velaei
- Department of Anatomical Science, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Balal Barazvan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleimani Rad
- Department of Anatomical Science, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Gallo-Oller G, Vollmann-Zwerenz A, Meléndez B, Rey JA, Hau P, Dotor J, Castresana JS. P144, a Transforming Growth Factor beta inhibitor peptide, generates antitumoral effects and modifies SMAD7 and SKI levels in human glioblastoma cell lines. Cancer Lett 2016; 381:67-75. [PMID: 27473823 DOI: 10.1016/j.canlet.2016.07.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 12/22/2022]
Abstract
Glioblastoma (GBM) is the most prevalent malignant primary brain tumor, accounting for 60-70% of all gliomas. Current median patient survival time is 14-16 months after diagnosis. Numerous efforts in therapy have not significantly altered the nearly uniform lethality of this malignancy. The Transforming Growth Factor beta (TGF-β) signaling pathway plays a key role in GBM and is implicated in proliferation, invasion and therapy resistance. Several inhibitors of the TGF-β pathway have entered clinical trials or are under development. In this work, the therapeutic potential of P144, a TGF-β inhibitor peptide, was analyzed. P144 decreased proliferation, migration, invasiveness, and tumorigenicity in vitro, whereas apoptosis and anoikis were significantly increased for GBM cell lines. SMAD2 phosphorylation was reduced, together with a downregulation of SKI and an upregulation of SMAD7 at both transcriptional and translational levels. Additionally, P144 was able to impair tumor growth and increase survival in an in vivo flank model. Our findings suggest a potential effect of P144 in vitro and in vivo that is mediated by regulation of transcriptional target genes of the TGF-β pathway, suggesting a therapeutic potential of P144 for GBM treatment.
Collapse
Affiliation(s)
- Gabriel Gallo-Oller
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
| | - Arabel Vollmann-Zwerenz
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Bárbara Meléndez
- Molecular Pathology Research Unit, Department of Pathology, Virgen de la Salud Hospital, Toledo, Spain
| | - Juan A Rey
- IdiPaz Research Unit, La Paz University Hospital, Madrid, Spain
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | | | - Javier S Castresana
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain.
| |
Collapse
|
17
|
Wang X, Gao Z, Wu X, Zhang W, Zhou G, Liu W. Inhibitory effect of TGF-β peptide antagonist on the fibrotic phenotype of human hypertrophic scar fibroblasts. PHARMACEUTICAL BIOLOGY 2016; 54:1189-1197. [PMID: 26135051 DOI: 10.3109/13880209.2015.1059862] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT TGF-β plays a central role in hypertrophic scar (HS) formation and development. OBJECTIVE This study investigated the role of a TGF-β antagonist peptide in inhibiting fibrotic behavior of human HS-derived fibroblasts (HSFs). MATERIALS AND METHODS HSFs were seeded at a density of 3.1 × 10(4)/cm(2) and were subjected to treatment of peptide antagonist (30 μM) or TGF-β receptor inhibitor LY2109761 (10 μM) or without treatment followed by the analyses of quantitative PCR, Elisa, in vitro wounding and fibroblast-populated collagen lattice (FPCL) assays. RESULTS qPCR and Elisa analyses showed that the peptide could, respectively, reduce the gene (at 48 h) and protein (at 72 h) expression levels of collagen I (86 ± 4.8%; 56.6 ± 7.3%), collagen III (73 ± 10.7%; 43.7 ± 7.2%), fibronectin (90 ± 8.9%; 21.1 ± 2.8%), and TGF-β1 (85 ± 9.3%; 25.0 ± 9.4%) as opposed to the non-treated group (p < 0.05), as the LY2109761 group similarly did. Cell proliferation was also significantly inhibited at day 5 (CCK-8 assay) by both peptide and LY2109761 treatments compared with the non-treated group (p < 0.05). The peptide also significantly inhibited cell migration as opposed to blank control at 24 h (43 ± 6.7% versus 60 ± 2.1%, p < 0.05) and at 48 h (63.9 ± 3.1% versus 95 ± 4.1%, p < 0.05). Similar to LY2109761, the peptide antagonist significantly reduced HS FPCL contraction compared with the non-treated group with significant differences in surface area at 48 h (0.71 ± 0.06 cm(2) versus 0.51 ± 0.06 cm(2), p < 0.05) and at 72 h (0.65 ± 0.02 cm(2) versus 0.42 ± 0.01 cm(2), p < 0.05). CONCLUSION The TGF-β antagonist peptide may serve as an important drug for HS prevention and reduction given the obvious benefits of good biosafety, low cost, and easy manufacture and delivery.
Collapse
Affiliation(s)
- Xiuxia Wang
- a Department of Plastic and Reconstructive Surgery , Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , PR China and
| | - Zhen Gao
- a Department of Plastic and Reconstructive Surgery , Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , PR China and
| | - Xiaoli Wu
- a Department of Plastic and Reconstructive Surgery , Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , PR China and
| | - Wenjie Zhang
- a Department of Plastic and Reconstructive Surgery , Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , PR China and
- b National Tissue Engineering Center of China , Shanghai , PR China
| | - Guangdong Zhou
- a Department of Plastic and Reconstructive Surgery , Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , PR China and
- b National Tissue Engineering Center of China , Shanghai , PR China
| | - Wei Liu
- a Department of Plastic and Reconstructive Surgery , Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , PR China and
- b National Tissue Engineering Center of China , Shanghai , PR China
| |
Collapse
|
18
|
Damdinsuren A, Matsushita H, Ito M, Tanaka M, Jin G, Tsukamoto H, Asai S, Ando K, Miyachi H. FLT3-ITD drives Ara-C resistance in leukemic cells via the induction of RUNX3. Leuk Res 2015; 39:1405-13. [DOI: 10.1016/j.leukres.2015.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 08/23/2015] [Accepted: 09/06/2015] [Indexed: 12/11/2022]
|
19
|
Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST, Cleverly AL, Desaiah D, Guba SC, Benhadji KA, Slapak CA, Lahn MM. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther 2015; 9:4479-99. [PMID: 26309397 PMCID: PMC4539082 DOI: 10.2147/dddt.s86621] [Citation(s) in RCA: 248] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) signaling regulates a wide range of biological processes. TGF-β plays an important role in tumorigenesis and contributes to the hallmarks of cancer, including tumor proliferation, invasion and metastasis, inflammation, angiogenesis, and escape of immune surveillance. There are several pharmacological approaches to block TGF-β signaling, such as monoclonal antibodies, vaccines, antisense oligonucleotides, and small molecule inhibitors. Galunisertib (LY2157299 monohydrate) is an oral small molecule inhibitor of the TGF-β receptor I kinase that specifically downregulates the phosphorylation of SMAD2, abrogating activation of the canonical pathway. Furthermore, galunisertib has antitumor activity in tumor-bearing animal models such as breast, colon, lung cancers, and hepatocellular carcinoma. Continuous long-term exposure to galunisertib caused cardiac toxicities in animals requiring adoption of a pharmacokinetic/pharmacodynamic-based dosing strategy to allow further development. The use of such a pharmacokinetic/pharmacodynamic model defined a therapeutic window with an appropriate safety profile that enabled the clinical investigation of galunisertib. These efforts resulted in an intermittent dosing regimen (14 days on/14 days off, on a 28-day cycle) of galunisertib for all ongoing trials. Galunisertib is being investigated either as monotherapy or in combination with standard antitumor regimens (including nivolumab) in patients with cancer with high unmet medical needs such as glioblastoma, pancreatic cancer, and hepatocellular carcinoma. The present review summarizes the past and current experiences with different pharmacological treatments that enabled galunisertib to be investigated in patients.
Collapse
Affiliation(s)
| | - J Scott Sawyer
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Anja J Stauber
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Kyla E Driscoll
- Lilly Research Laboratories, Eli Lilly and Company, New York, NY, USA
| | - Shawn T Estrem
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Ann L Cleverly
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Durisala Desaiah
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Susan C Guba
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Karim A Benhadji
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Michael M Lahn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
20
|
Tanaka K, Miyata H, Sugimura K, Fukuda S, Kanemura T, Yamashita K, Miyazaki Y, Takahashi T, Kurokawa Y, Yamasaki M, Wada H, Nakajima K, Takiguchi S, Mori M, Doki Y. miR-27 is associated with chemoresistance in esophageal cancer through transformation of normal fibroblasts to cancer-associated fibroblasts. Carcinogenesis 2015; 36:894-903. [PMID: 26026166 DOI: 10.1093/carcin/bgv067] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 04/20/2015] [Indexed: 02/07/2023] Open
Abstract
There is increasing evidence that the expression of microRNA (miRNA) in cancer is associated with chemosensitivity but the mechanism of miRNA-induced chemoresistance has not been fully elucidated. The aim of this study was to examine the role of extracellular miRNA in the response to chemotherapy in esophageal cancer. First, serum expression of miRNAs selected by miRNA array was measured by quantitative reverse transcription-polymerase chain reaction in 68 patients with esophageal cancer who received cisplatin-based chemotherapy to examine the relationship between miRNA expression and response to chemotherapy. The serum expression levels of 18 miRNAs were different between responders and non-responders by miRNA array. Of these, high expression levels of miR-27a/b correlated with poor response to chemotherapy in patients with esophageal cancer. Next, in vitro assays were conducted to investigate the mechanism of miRNA-induced chemoresistance. Although transfection of miR-27a/b to cancer cells had no significant impact on chemosensitivity, esophageal cancer cells cultured in supernatant of miR-27a/b-transfected normal fibroblast showed reduced chemosensitivity to cisplatin, compared with cancer cells cultured in supernatant of normal fibroblast. MiR-27a/b-transfected normal fibroblast showed α-smooth muscle actin (α-SMA) expression, a marker of cancer-associated fibroblasts (CAF) and increased production of transforming growth factor-β (TGF-β). Chemosensitivity recovered after administration of neutralizing antibody of TGF-β to the supernatant transfer experiments. Our results indicated that miR-27a/b is involved in resistance to chemotherapy in esophageal cancer, through miR-27a/b-induced transformation of normal fibroblast into CAF.
Collapse
Affiliation(s)
- Koji Tanaka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Hiroshi Miyata
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Keijiro Sugimura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Shuichi Fukuda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Takashi Kanemura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Kotaro Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Yasuhiro Miyazaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Hisashi Wada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita city, Osaka 565-0871, Japan
| |
Collapse
|
21
|
Dittmer J, Leyh B. The impact of tumor stroma on drug response in breast cancer. Semin Cancer Biol 2014; 31:3-15. [PMID: 24912116 DOI: 10.1016/j.semcancer.2014.05.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/27/2014] [Accepted: 05/30/2014] [Indexed: 02/06/2023]
Abstract
In the last two decades the breast cancer mortality rate has steadily declined, in part, due to the availability of better treatment options. However, drug resistance still remains a major challenge. Resistance can be an inherent feature of breast cancer cells, but can also arise from the tumor microenvironment. This review aims to focus on the modulatory effect of the tumor microenvironment on the differing response of breast cancer subtypes to targeted drugs and chemotherapy.
Collapse
Affiliation(s)
- Jürgen Dittmer
- Clinic for Gynecology, University of Halle, Halle/Saale, Germany.
| | - Benjamin Leyh
- Clinic for Gynecology, University of Halle, Halle/Saale, Germany
| |
Collapse
|
22
|
Castells M, Milhas D, Gandy C, Thibault B, Rafii A, Delord JP, Couderc B. Microenvironment mesenchymal cells protect ovarian cancer cell lines from apoptosis by inhibiting XIAP inactivation. Cell Death Dis 2013; 4:e887. [PMID: 24176845 PMCID: PMC3824693 DOI: 10.1038/cddis.2013.384] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 08/23/2013] [Accepted: 08/26/2013] [Indexed: 01/29/2023]
Abstract
Epithelial ovarian carcinoma is characterized by high frequency of recurrence (70% of patients) and carboplatin resistance acquisition. Carcinoma-associated mesenchymal stem cells (CA-MSC) have been shown to induce ovarian cancer chemoresistance through trogocytosis. Here we examined CA-MSC properties to protect ovarian cancer cells from carboplatin-induced apoptosis. Apoptosis was determined by Propidium Iodide and Annexin-V-FITC labelling and poly-ADP-ribose polymerase cleavage analysis. We showed a significant increase of inhibitory concentration 50 and a 30% decrease of carboplatin-induced apoptosis in ovarian cancer cells incubated in the presence of CA-MSC-conditioned medium (CM). A molecular analysis of apoptosis signalling pathway in response to carboplatin revealed that the presence of CA-MSC CM induced a 30% decrease of effector caspases-3 and -7 activation and proteolysis activity. CA-MSC secretions promoted Akt and X-linked inhibitor of apoptosis protein (XIAP; caspase inhibitor from inhibitor of apoptosis protein (IAP) family) phosphorylation. XIAP depletion by siRNA strategy permitted to restore apoptosis in ovarian cancer cells stimulated by CA-MSC CM. The factors secreted by CA-MSC are able to confer chemoresistance to carboplatin in ovarian cancer cells through the inhibition of effector caspases activation and apoptosis blockade. Activation of the phosphatidylinositol 3-kinase (PI3K)/Akt signalling pathway and the phosphorylation of its downstream target XIAP underlined the implication of this signalling pathway in ovarian cancer chemoresistance. This study reveals the potentialities of targeting XIAP in ovarian cancer therapy.
Collapse
Affiliation(s)
- M Castells
- 1] EA4553, Institut Claudius Regaud, Toulouse F-31052, France [2] University of Toulouse III, Toulouse F-31062, France
| | | | | | | | | | | | | |
Collapse
|
23
|
Tabe Y, Shi YX, Zeng Z, Jin L, Shikami M, Hatanaka Y, Miida T, Hsu FJ, Andreeff M, Konopleva M. TGF-β-Neutralizing Antibody 1D11 Enhances Cytarabine-Induced Apoptosis in AML Cells in the Bone Marrow Microenvironment. PLoS One 2013; 8:e62785. [PMID: 23826077 PMCID: PMC3695026 DOI: 10.1371/journal.pone.0062785] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 03/25/2013] [Indexed: 01/10/2023] Open
Abstract
Hypoxia and interactions with bone marrow (BM) stromal cells have emerged as essential components of the leukemic BM microenvironment in promoting leukemia cell survival and chemoresistance. High levels of transforming growth factor beta 1 (TGFβ1) produced by BM stromal cells in the BM niche regulate cell proliferation, survival, and apoptosis, depending on the cellular context. Exogenous TGFβ1 induced accumulation of acute myeloid leukemia (AML) cells in a quiescent G0 state, which was further facilitated by the co-culture with BM-derived mesenchymal stem cells (MSCs). In turn, TGFβ-neutralizing antibody 1D11 abrogated rhTGFβ1 induced cell cycle arrest. Blocking TGFβ with 1D11 further enhanced cytarabine (Ara-C)-induced apoptosis of AML cells in hypoxic and in normoxic conditions. Additional constituents of BM niche, the stroma-secreted chemokine CXCL12 and its receptor CXCR4 play crucial roles in cell migration and stroma/leukemia cell interactions. Treatment with 1D11 combined with CXCR4 antagonist plerixafor and Ara-C decreased leukemia burden and prolonged survival in an in vivo leukemia model. These results indicate that blockade of TGFβ by 1D11 and abrogation of CXCL12/CXCR4 signaling may enhance the efficacy of chemotherapy against AML cells in the hypoxic BM microenvironment.
Collapse
Affiliation(s)
- Yoko Tabe
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Clinical Laboratory Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Yue Xi Shi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Zhihong Zeng
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Linhua Jin
- Department of Clinical Laboratory Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Masato Shikami
- Department of Hematology, Aichi Medical University, Aichi, Japan
| | - Yasuhito Hatanaka
- Department of Clinical Laboratory Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Takashi Miida
- Department of Clinical Laboratory Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Frank J. Hsu
- Genzyme Corporation, Cambridge, Massachusetts, United States of America
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
24
|
Fang Y, Chen Y, Yu L, Zheng C, Qi Y, Li Z, Yang Z, Zhang Y, Shi T, Luo J, Liu M. Inhibition of breast cancer metastases by a novel inhibitor of TGFβ receptor 1. J Natl Cancer Inst 2012. [PMID: 23178439 DOI: 10.1093/jnci/djs485] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Transforming growth factor beta (TGFβ), which is implicated in metastasis to various organs in breast cancer, is a potential target for new antitumor metastasis drugs. METHODS To identify specific inhibitors of TGFβ receptor 1 (TGFβR1) in breast cancer metastasis, a virtual library of more than 400000 different compounds was screened by molecular docking modeling and confirmed with Smad-binding element luciferase and TGFβR1 kinase assays. Affymetrix GeneChip expression analysis of mRNA levels and real-time polymerase chain reaction were performed to determine expression changes of TGFβ-mediated, metastasis-associated genes in breast cancer cells after treatment with the small-molecule inhibitor YR-290. YR-290 was also examined for its effects on breast cancer migration, invasion, and metastasis using transwell and epithelial-to-mesenchymal transition (EMT) assays in vitro and three different mouse (BALB/c and NU/NU nude) models (n = 10 per group). Kaplan-Meier analyses were used to assess survival. All statistical tests were two-sided. RESULTS YR-290 interacted with the kinase domain of TGFβR1, abrogated kinase activity (half maximal inhibitory concentration = 137nM, 95% confidence interval = 126.4 to 147.6nM) and inhibited the TGFβ-mediated downstream signaling pathway and metastasis-associated genes in breast cancer cells. YR-290 inhibited TGFβ-modulated breast cancer cell migration and invasion. In tumor metastasis mouse models, YR-290 almost completely blocked cancer metastasis. Numbers of lung tumor nodules of mice treated with 1mg/kg and 5mg/kg YR-290 were reduced by 74.93% (95% confidence interval = 61.45% to 88.41%) and 94.93% (95% confidence interval = 82.13% to 100%), respectively, compared with control mice. Treatment with YR-290 also statistically significantly prolonged the survival of tumor-bearing mice. CONCLUSIONS YR-290 is a novel inhibitor of tumor metastasis that works by blocking TGFβ signaling pathways.
Collapse
Affiliation(s)
- Yuanzhang Fang
- Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Rd, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Castells M, Thibault B, Delord JP, Couderc B. Implication of tumor microenvironment in chemoresistance: tumor-associated stromal cells protect tumor cells from cell death. Int J Mol Sci 2012; 13:9545-9571. [PMID: 22949815 PMCID: PMC3431813 DOI: 10.3390/ijms13089545] [Citation(s) in RCA: 186] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/13/2012] [Accepted: 07/17/2012] [Indexed: 12/17/2022] Open
Abstract
Tumor development principally occurs following the accumulation of genetic and epigenetic alterations in tumor cells. These changes pave the way for the transformation of chemosensitive cells to chemoresistant ones by influencing the uptake, metabolism, or export of drugs at the cellular level. Numerous reports have revealed the complexity of tumors and their microenvironment with tumor cells located within a heterogeneous population of stromal cells. These stromal cells (fibroblasts, endothelial or mesothelial cells, adipocytes or adipose tissue-derived stromal cells, immune cells and bone marrow-derived stem cells) could be involved in the chemoresistance that is acquired by tumor cells via several mechanisms: (i) cell-cell and cell-matrix interactions influencing the cancer cell sensitivity to apoptosis; (ii) local release of soluble factors promoting survival and tumor growth (crosstalk between stromal and tumor cells); (iii) direct cell-cell interactions with tumor cells (crosstalk or oncologic trogocytosis); (iv) generation of specific niches within the tumor microenvironment that facilitate the acquisition of drug resistance; or (v) conversion of the cancer cells to cancer-initiating cells or cancer stem cells. This review will focus on the implication of each member of the heterogeneous population of stromal cells in conferring resistance to cytotoxins and physiological mediators of cell death.
Collapse
Affiliation(s)
| | | | | | - Bettina Couderc
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +33-5-61-42-46-44; Fax: +33-5-61-42-46-31
| |
Collapse
|
26
|
Regulation of C/EBPβ and resulting functions in cells of the monocytic lineage. Cell Signal 2012; 24:1287-96. [DOI: 10.1016/j.cellsig.2012.02.007] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 02/14/2012] [Indexed: 01/10/2023]
|
27
|
Giannelli G, Mazzocca A, Fransvea E, Lahn M, Antonaci S. Inhibiting TGF-β signaling in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2011; 1815:214-23. [PMID: 21129443 DOI: 10.1016/j.bbcan.2010.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 11/18/2010] [Accepted: 11/20/2010] [Indexed: 12/17/2022]
|
28
|
Abstract
INTRODUCTION Integrins, which are heterodimeric membrane glycoproteins, consist of a family of cell-surface receptors mediating cell-matrix and cell-cell adhesion. Analysis of tumor-associated integrins has revealed an important relationship between integrins and tumor development, bringing new insights into integrin-based cancer therapies. Hepatocellular carcinoma (HCC) is one of the most malignant tumors worldwide and integrins appeal to be a novel group of potential therapeutic targets for HCC. AREAS COVERED This review summarizes the current knowledge of integrins involved in HCC and the potential of integrin-targeted drugs in HCC therapy. A brief introduction on the structure, biological function and regulatory mechanism of integrins is given. The distinct expression patterns and biological functions of HCC-associated integrins are described. Finally, the current situation of integrin-based therapies in HCC and other tumor types are extensively discussed in the light of their implications in preclinical and clinical trials. EXPERT OPINION To date, increasing numbers of integrin-targeted drugs are undergoing development and they exhibit diverse effects in cancer clinical trials. Tumor heterogeneity should be emphasized in developing effective integrin-targeted drugs specific for HCC. A better understanding of how integrins cooperatively function in HCC will assist in designing more successful integrin-targeted therapeutic drugs and corresponding approaches.
Collapse
Affiliation(s)
- Yanhua Wu
- Fudan University, Institute of Genetics, State Key Laboratory of Genetic Engineering, 220 Handan Road, Shanghai, 200433, P. R. China
| | | | | | | |
Collapse
|
29
|
Otten J, Bokemeyer C, Fiedler W. Tgf-Beta superfamily receptors-targets for antiangiogenic therapy? JOURNAL OF ONCOLOGY 2010; 2010:317068. [PMID: 20490264 PMCID: PMC2871186 DOI: 10.1155/2010/317068] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 02/23/2010] [Indexed: 01/17/2023]
Abstract
The TGF-beta pathway controls a broad range of cellular behavior including cell proliferation, differentiation, and apoptosis of various cell types including tumor cells, endothelial cells, immune cells, and fibroblasts. Besides TGF-beta's direct effects on tumor growth and its involvement in neoangiogenesis have received recent attention. Germline mutations in TGF-beta receptors or coreceptors causing Hereditary Hemorrhagic Teleangiectasia and the Loeys-Dietz syndrome underline the involvement of TGF-beta in vessel formation and maturation. Several therapeutic approaches are evaluated at present targeting the TGF-beta pathway including utilization of antisense oligonucleotides against TGF-beta itself or antibodies or small molecule inhibitors of TGF-beta receptors. Some of these therapeutic agents have already entered the clinical arena including an antibody against the endothelium specific TGF-beta class I receptor ALK-1 targeting tumor vasculature. In conclusion, therapeutic manipulation of the TGF-beta pathway opens great opportunities in future cancer therapy.
Collapse
Affiliation(s)
- Jasmin Otten
- Sections of Pneumonology and Bone Marrow Transplantation, Department of Oncology and Hematology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Carsten Bokemeyer
- Sections of Pneumonology and Bone Marrow Transplantation, Department of Oncology and Hematology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Walter Fiedler
- Sections of Pneumonology and Bone Marrow Transplantation, Department of Oncology and Hematology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
30
|
Døsen-Dahl G, Munthe E, Nygren MK, Stubberud H, Hystad ME, Rian E. Bone marrow stroma cells regulate TIEG1 expression in acute lymphoblastic leukemia cells: Role of TGFβ/BMP-6 and TIEG1 in chemotherapy escape. Int J Cancer 2008; 123:2759-66. [DOI: 10.1002/ijc.23833] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|