1
|
Zhang W, Cao X, Wu H, Zhong X, Shi Y, Sun Z. Function of Steroid Receptor Coactivators in T Cells and Cancers: Implications for Cancer Immunotherapy. Crit Rev Immunol 2024; 44:111-126. [PMID: 38848298 PMCID: PMC11902286 DOI: 10.1615/critrevimmunol.2024051613] [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] [Indexed: 06/09/2024]
Abstract
Steroid receptor coactivator (SRC) family members (SRC1, SRC2 and SRC3) are transcriptional co-regulators. SRCs orchestrate gene transcription by inducing transactivation of nuclear receptors and other transcription factors. Overexpression of SRCs is widely implicated in a range of cancers, especially hormone-related cancers. As coactivators, SRCs regulate multiple metabolic pathways involved in tumor growth, invasion, metastasis, and chemo-resistance. Emerging evidence in recent years suggest that SRCs also regulate maturation, differentiation, and cytotoxicity of T cells by controlling metabolic activities. In this review, we summarize the current understanding of the function of SRCs in T cells as well as cancer cells. Importantly, the controversies of targeting SRCs for cancer immunotherapy as well as possible reconciliation strategies are also discussed.
Collapse
Affiliation(s)
- Wencan Zhang
- Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Hongmin Wu
- Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Xiancai Zhong
- Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Yun Shi
- Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Zuoming Sun
- Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| |
Collapse
|
2
|
Liao C, Liu Y, Lin Y, Wang J, Zhou T, Weng W. Mesenchymal Stem Cell-conditioned Medium Protecting Renal Tubular Epithelial Cells by Inhibiting Hypoxia-inducible Factor-1α and Nuclear Receptor Coactivator-1. Curr Stem Cell Res Ther 2024; 19:1369-1381. [PMID: 37817516 DOI: 10.2174/011574888x247652230928064627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Acute kidney injury (AKI) is characterized by inflammatory infiltration and damage and death of renal tubular epithelial cells (RTECs), in which hypoxia plays an important role. Deferoxamine (DFO) is a well-accepted chemical hypoxia-mimetic agent. Mesenchymal stem cell-conditioned medium (MSC-CM) can reduce local inflammation and repair tissue. In this study, we explored the effect and molecular mechanism of MSC-CM-mediated protection of RTECs under DFO-induced hypoxia. METHODS Rat renal proximal tubule NRK-52E cells were treated with different concentrations of DFO for 24 hours, followed by evaluation of RTEC injury, using a Cell Counting Kit-8 (CCK-8) to detect cell viability and western blotting to evaluate the expression of transforming growth factor- beta 1 (TGF-β1), α-smooth muscle actin (α-SMA), and hypoxia-inducible factor-1 alpha (HIF-1α) in NRK-52E cells. Then, three groups of NRK-52E cells were used in experiments, including normal control (NC), 25 μM DFO, and 25 μM DFO + MSC-CM. MSC-CM was obtained from the human umbilical cord. MSC-CM was used to culture cells for 12 hours before DFO treatment, then fresh MSC-CM and 25 μM DFO were added, and cells were cultured for another 24 hours before analysis. RESULTS Western blotting and cellular immunofluorescence staining showed culture of NRK-52E cells in 25 μM DFO for 24 hours induced HIF-1α and nuclear receptor coactivator-1 (NCoA-1), simulating hypoxia. MSC-CM could inhibit the DFO-induced up-regulation of α-SMA, TGF-β1, HIF-1α and NCoA-1. CONCLUSION Our results suggest that MSC-CM has a protective effect on RTECs by down-regulating HIF-1α and NCoA-1, which may be the harmful factors in renal injury.
Collapse
Affiliation(s)
- Chunling Liao
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yiping Liu
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yongda Lin
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Jiali Wang
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Wenjuan Weng
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| |
Collapse
|
3
|
Zhang J, Pan L, Zhang S, Yang Y, Liang J, Ma S, Wu Q. CISD2 promotes lung squamous carcinoma cell migration and invasion via the TGF-β1-induced Smad2/3 signaling pathway. Clin Transl Oncol 2023; 25:3527-3540. [PMID: 37249759 DOI: 10.1007/s12094-023-03222-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/19/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Although aberrant expression of CDGSH iron sulfur domain 2 (CISD2) contributes to the tumorigenesis and progression of numerous human cancers, the biological function of CISD2 and its specific prognostic value in lung squamous cell carcinoma (LUSC) have yet to be comprehensively explored. The current study aimed to elucidate the role of CISD2 in LUSC as well as the underlying molecular mechanisms. METHODS Immunohistochemistry was conducted to detect the protein expression of CISD2 and analyze whether high expression of CISD2 affects the overall survival (OS) of LUSC patients. Cell proliferation, colony formation, wound healing and Transwell invasion assays were performed to clarify whether CISD2 contributes to LUSC cell proliferation and disease progression. Quantitative real-time reverse transcription-PCR and western blot assays were used to detect the levels of transcription factors and key epithelial-mesenchymal transition (EMT)-related markers in LUSC cells after CISD2 knockdown and overexpression to determine whether CISD2 regulates transforming growth factor-beta (TGF-β)-induced EMT in LUSC. RESULTS Immunohistochemistry of human tissue microarrays containing 90 pairs of adjacent and cancerous tissues revealed that CISD2 is considerably overexpressed in LUSC and strongly linked to poor OS. Functional experiments suggested that silencing endogenous CISD2 inhibited the growth, colony formation, migration, and invasion of H2170 and H226 cell lines. Exogenous overexpression of CISD2 facilitated these phenotypes in SK-MES-1 and H2170 cells. Furthermore, CISD2 promoted EMT progression by increasing the expression of mesenchymal markers (N-cadherin, vimentin, Snail, and Slug) as well as SMAD2/3 and reducing the expression of the epithelial marker E-cadherin. Mechanistically, our studies provide the first evidence that CISD2 can promote EMT by enhancing TGF-β1-induced Smad2/3 expression in LUSC cells. CONCLUSION In conclusion, our research illustrates that CISD2 is highly expressed in LUSC and may facilitate LUSC proliferation and metastasis. Thus, CISD2 may serve as an independent prognostic marker and possible treatment target for LUSC.
Collapse
Affiliation(s)
- Jingjing Zhang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lifang Pan
- Integrated Traditional Chinese and Western Medicine Oncology Laboratory, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Shirong Zhang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yuhong Yang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jiafeng Liang
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Shenglin Ma
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China.
| | - Qiong Wu
- Integrated Traditional Chinese and Western Medicine Oncology Laboratory, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| |
Collapse
|
4
|
Spatial-Temporal Patterns and Inflammatory Factors of Bone Matrix Remodeling. Stem Cells Int 2021; 2021:4307961. [PMID: 34777503 PMCID: PMC8580647 DOI: 10.1155/2021/4307961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 11/18/2022] Open
Abstract
The bone extracellular matrix (ECM) contains organic and mineral constituents. The establishment and degradation processes of ECM connect with spatial and temporal patterns, especially circadian rhythms in ECM. These patterns are responsible for the physical and biological characteristics of bone. The disturbances of the patterns disrupt bone matrix remodeling and cause diverse bone diseases, such as osteogenesis imperfecta (OI) and bone fracture. In addition, the main regulatory factors and inflammatory factors also follow circadian rhythms. Studies show that the circadian oscillations of these factors in bone ECM potentially influence the interactions between immune responses and bone formation. More importantly, mesenchymal stem cells (MSCs) within the specific microenvironments provide the regenerative potential for tissue remodeling. In this review, we summarize the advanced ECM spatial characteristics and the periodic patterns of bone ECM. Importantly, we focus on the intrinsic connections between the immunoinflammatory system and bone formation according to circadian rhythms of regulatory factors in bone ECM. And our research group emphasizes the multipotency of MSCs with their microenvironments. The advanced understandings of bone ECM formation patterns and MSCs contribute to providing optimal prevention and treatment strategies.
Collapse
|
5
|
Huang Y, Duan X, Wang Z, Sun Y, Guan Q, Kang L, Zhang Q, Fang L, Li J, Wong J. An acetylation-enhanced interaction between transcription factor Sox2 and the steroid receptor coactivators facilitates Sox2 transcriptional activity and function. J Biol Chem 2021; 297:101389. [PMID: 34762910 PMCID: PMC8668987 DOI: 10.1016/j.jbc.2021.101389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/02/2022] Open
Abstract
SRY-box 2 (Sox2) is a transcription factor with critical roles in maintaining embryonic stem (ES) cell and adult stem cell functions and in tumorigenesis. However, how Sox2 exerts its transcriptional function remains unclear. Here, we used an in vitro protein–protein interaction assay to discover transcriptional regulators for ES cell core transcription factors (Oct4, Sox2, Klf4, and c-Myc) and identified members of the steroid receptor coactivators (SRCs) as Sox2-specific interacting proteins. The SRC family coactivators have broad roles in transcriptional regulation, but it is unknown whether they also serve as Sox2 coactivators. We demonstrated that these proteins facilitate Sox2 transcriptional activity and act synergistically with p300. Furthermore, we uncovered an acetylation-enhanced interaction between Sox2 and SRC-2/3, but not SRC-1, demonstrating it is Sox2 acetylation that promotes the interaction. We identified putative Sox2 acetylation sites required for acetylation-enhanced interaction between Sox2 and SRC-3 and demonstrated that acetylation on these sites contributes to Sox2 transcriptional activity and recruitment of SRC-3. We showed that activation domains 1 and 2 of SRC-3 both display a preferential binding to acetylated Sox2. Finally, functional analyses in mouse ES cells demonstrated that knockdown of SRC-2/3 but not SRC-1 in mouse ES cells significantly downregulates the transcriptional activities of various Sox2 target genes and impairs ES cell stemness. Taken together, we identify specific SRC family proteins as novel Sox2 coactivators and uncover the role of Sox2 acetylation in promoting coactivator recruitment and Sox2 transcriptional function.
Collapse
Affiliation(s)
- Yuanyong Huang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiaoya Duan
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhen Wang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yimei Sun
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Qingqing Guan
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Li Kang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Qiao Zhang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Lan Fang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiwen Li
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China; Joint Center for Translational Medicine, Fengxian District Central Hospital, Shanghai, China.
| |
Collapse
|
6
|
Gao J, Ye J, Ying Y, Lin H, Luo Z. Negative regulation of TGF-β by AMPK and implications in the treatment of associated disorders. Acta Biochim Biophys Sin (Shanghai) 2018; 50:523-531. [PMID: 29873702 DOI: 10.1093/abbs/gmy028] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Indexed: 01/18/2023] Open
Abstract
Transforming growth factor beta (TGF-β) regulates a large number of biological processes, including proliferation, differentiation, immune response, and development. In addition, TGF-β plays important roles in some pathological processes, for instance, it is upregulated and activated in fibrosis and advanced cancer. Adenosine monophosphate-activated protein kinase (AMPK) acts as a fuel gauge that is activated when cells sense shortage of ATP and increase in AMP or AMP:ATP ratio. Activation of AMPK slows down anabolic processes and stimulates catabolic processes, leading to increased production of ATP. Furthermore, the functions of AMPK have been extended beyond energy homeostasis. In fact, AMPK has been shown to exert a tumor suppressive effect. Recent studies have demonstrated negative impacts of AMPK on TGF-β function. Therefore, in this review, we will discuss the differences in the biological functions of TGF-β and AMPK, and some pathological processes such as fibrosis, epithelial-mesenchymal transition (EMT) and cancer metastasis, as well as angiogenesis and heterotopic ossifications where TGF-β and AMPK exert opposite effects.
Collapse
Affiliation(s)
- Jiayu Gao
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Jiangxi Medical College, Nanchang 330000, China
- Department of Pathology, Schools of Basic Sciences, Nanchang University Jiangxi Medical College, Nanchang 330000, China
| | - Jinhui Ye
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Jiangxi Medical College, Nanchang 330000, China
| | - Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Jiangxi Medical College, Nanchang 330000, China
| | - Hui Lin
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Jiangxi Medical College, Nanchang 330000, China
| | - Zhijun Luo
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Jiangxi Medical College, Nanchang 330000, China
- Department of Pathology, Schools of Basic Sciences, Nanchang University Jiangxi Medical College, Nanchang 330000, China
| |
Collapse
|
7
|
Salter RC, Foka P, Davies TS, Gallagher H, Michael DR, Ashlin TG, Ramji DP. The role of mitogen-activated protein kinases and sterol receptor coactivator-1 in TGF-β-regulated expression of genes implicated in macrophage cholesterol uptake. Sci Rep 2016; 6:34368. [PMID: 27687241 PMCID: PMC5043369 DOI: 10.1038/srep34368] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 09/12/2016] [Indexed: 01/25/2023] Open
Abstract
The anti-atherogenic cytokine TGF-β inhibits macrophage foam cell formation by suppressing the expression of key genes implicated in the uptake of modified lipoproteins. We have previously shown a critical role for p38 MAPK and JNK in the TGF-β-mediated regulation of apolipoprotein E expression in human monocytes. However, the roles of these two MAPK pathways in the control of expression of key genes involved in the uptake of modified lipoproteins in human macrophages is poorly understood and formed the focus of this study. TGF-β activated both p38 MAPK and JNK, and knockdown of p38 MAPK or c-Jun, a key downstream target of JNK action, demonstrated their requirement in the TGF-β-inhibited expression of several key genes implicated in macrophage lipoprotein uptake. The potential role of c-Jun and specific co-activators in the action of TGF-β was investigated further by studies on the lipoprotein lipase gene. c-Jun did not directly interact with the minimal promoter region containing the TGF-β response elements and a combination of transient transfection and knock down assays revealed an important role for SRC-1. These studies provide novel insights into the mechanisms underlying the TGF-β-mediated inhibition of macrophage gene expression associated with the control of cholesterol homeostasis.
Collapse
Affiliation(s)
- Rebecca C Salter
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX, United Kingdom
| | - Pelagia Foka
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX, United Kingdom
| | - Thomas S Davies
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX, United Kingdom
| | - Hayley Gallagher
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX, United Kingdom
| | - Daryn R Michael
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX, United Kingdom
| | - Tim G Ashlin
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX, United Kingdom
| | - Dipak P Ramji
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX, United Kingdom
| |
Collapse
|
8
|
Rouce RH, Shaim H, Sekine T, Weber G, Ballard B, Ku S, Barese C, Murali V, Wu MF, Liu H, Shpall EJ, Bollard CM, Rabin KR, Rezvani K. The TGF-β/SMAD pathway is an important mechanism for NK cell immune evasion in childhood B-acute lymphoblastic leukemia. Leukemia 2016; 30:800-11. [PMID: 26621337 PMCID: PMC4823160 DOI: 10.1038/leu.2015.327] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 09/28/2015] [Accepted: 10/15/2015] [Indexed: 01/05/2023]
Abstract
Natural killer (NK) cells are key components of the innate immune system, providing potent antitumor immunity. Here, we show that the tumor growth factor-β (TGF-β)/SMAD signaling pathway is an important mechanism for NK cell immune evasion in childhood B-acute lymphoblastic leukemia (ALL). We characterized NK cells in 50 consecutive children with B-ALL at diagnosis, end induction and during maintenance therapy compared with age-matched controls. ALL-NK cells at diagnosis had an inhibitory phenotype associated with impaired function, most notably interferon-γ production and cytotoxicity. By maintenance therapy, these phenotypic and functional abnormalities partially normalized; however, cytotoxicity against autologous blasts remained impaired. We identified ALL-derived TGF-β1 to be an important mediator of leukemia-induced NK cell dysfunction. The TGF-β/SMAD signaling pathway was constitutively activated in ALL-NK cells at diagnosis and end induction when compared with healthy controls and patients during maintenance therapy. Culture of ALL blasts with healthy NK cells induced NK dysfunction and an inhibitory phenotype, mediated by activation of the TGF-β/SMAD signaling pathway, and abrogated by blocking TGF-β. These data indicate that by regulating the TGF-β/SMAD pathway, ALL blasts induce changes in NK cells to evade innate immune surveillance, thus highlighting the importance of developing novel therapies to target this inhibitory pathway and restore antileukemic cytotoxicity.
Collapse
Affiliation(s)
- Rayne H. Rouce
- Texas Children’s Cancer and Hematology Centers/Baylor College of Medicine, Houston
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Hila Shaim
- Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston
| | - Takuya Sekine
- Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston
| | - Gerrit Weber
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Brandon Ballard
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Stephanie Ku
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Cecilia Barese
- Program for Cell Enhancement and Technologies for Immunotherapy, and Center for Cancer and Immunology Research, Children’s National Health System, Washington, DC
| | - Vineeth Murali
- Texas Children’s Cancer and Hematology Centers/Baylor College of Medicine, Houston
| | - Meng-Fen Wu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Hao Liu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Elizabeth J. Shpall
- Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston
| | - Catherine M. Bollard
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
- Program for Cell Enhancement and Technologies for Immunotherapy, and Center for Cancer and Immunology Research, Children’s National Health System, Washington, DC
| | - Karen R. Rabin
- Texas Children’s Cancer and Hematology Centers/Baylor College of Medicine, Houston
| | - Katayoun Rezvani
- Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston
| |
Collapse
|
9
|
Chen YT, Liao WL, Lin YJ, Chen SY, Tsai FJ. Association between SRC-1 gene polymorphisms and coronary artery aneurysms formation in Taiwanese children with Kawasaki disease. J Clin Lab Anal 2014; 28:435-9. [PMID: 24652666 DOI: 10.1002/jcla.21706] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Accepted: 10/02/2013] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Kawasaki disease (KD) patients who experience a cardiovascular complication known as a coronary artery aneurysm (CAA) are at high risk of developing ischemic heart disease, which may lead to sudden death. The etiology of CAA in KD patients is unclear, and this study aims to clarify the relationship between steroid receptor coactivator-1 (SRC-1) gene polymorphisms and CAA pathogenesis. METHODS We investigated four SRC-1 gene polymorphisms (rs11894248, rs17791703, rs7572475, and rs9309308) and their correlation with KD with CAA susceptibility in 327 Taiwanese people (279 KD patients without CAA and 48 KD patients with CAA). RESULTS The results indicated a statistically significant difference in genotype and allele frequency distributions at the SRC-1 four single nucleotide polymorphisms (SNPs) between KD patients with and without CAA (P < 0.01). Additionally, Smad3 gene polymorphism (rs12901071) is well known to be associated with KD patients. In our results, Smad3 SNP did not provide a statistically significant difference between KD patients with and without CAA. CONCLUSION Our data show that SRC-1 polymorphisms may be the underlying cause of CAA; therefore, the polymorphisms examined in this study warrant further investigation.
Collapse
Affiliation(s)
- Yng-Tay Chen
- Human Genetic Center, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | |
Collapse
|
10
|
Kim JI, Jung KJ, Jang HS, Park KM. Gender-specific role of HDAC11 in kidney ischemia- and reperfusion-induced PAI-1 expression and injury. Am J Physiol Renal Physiol 2013; 305:F61-70. [DOI: 10.1152/ajprenal.00015.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Male gender and the male hormone testosterone increase susceptibility to kidney ischemia and reperfusion (I/R) injury, which is associated with inflammatory responses. Possible involvement of histone deacetylase (HDAC) in inflammatory responses has been suggested. We investigated the gender-specific role of HDACs in plasminogen activator inhibitor type-1 (PAI-1) expression and I/R injury. PAI-1 inhibition protected the kidney from I/R-induced inflammation and functional loss. Among HDACs, only HDAC11 negatively regulated PAI-1 expression in I/R-subjected kidney gender specifically and lipopolysaccharide (LPS)-stimulated mouse monocytes/macrophages. HDAC11 gene silencing increased PAI-1 expression. Chromatin immunoprecipitation assay confirmed binding of HDAC11 to the promoter region of PAI-1 and then release by I/R insult or LPS treatment. I/R-induced HDAC11 release was inhibited by orchiectomy and reversed by dihydrotestosterone treatment. Release of HDAC11 increased acetylation of histone H3. In conclusion, male gender and male hormones accelerate I/R-induced decreases in expression and binding of HDAC11, resulting in an increase in PAI-1 expression. These data provide important insight into gender dimorphism offering HDAC11 as a novel target for I/R injury.
Collapse
Affiliation(s)
- Jee In Kim
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
- Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Kyong-Jin Jung
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
| | - Hee-Seong Jang
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
| | - Kwon Moo Park
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
- Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
11
|
Schiller M, Dennler S, Anderegg U, Kokot A, Simon JC, Luger TA, Mauviel A, Böhm M. Increased cAMP levels modulate transforming growth factor-beta/Smad-induced expression of extracellular matrix components and other key fibroblast effector functions. J Biol Chem 2009; 285:409-21. [PMID: 19858184 DOI: 10.1074/jbc.m109.038620] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
cAMP is a key messenger of many hormones and neuropeptides, some of which modulate the composition of extracellular matrix. Treatment of human dermal fibroblasts with dibutyryl cyclic AMP and forskolin antagonized the inductive effects of transforming growth factor-beta (TGF-beta) on the expression of collagen, connective tissue growth factor, tissue inhibitor of matrix metalloproteinase-1, and plasminogen activator inhibitor type I, four prototypical TGF-beta-responsive genes. Increased intracellular cAMP prevented TGF-beta-induced Smad-specific gene transactivation, although TGF-beta-mediated Smad phosphorylation and nuclear translocation remained unaffected. However, increased cAMP levels abolished TGF-beta-induced interaction of Smad3 with its transcriptional co-activator cAMP-response element-binding protein (CREB)-binding protein (CBP)/p300. Overexpression of the transcriptional co-activator CBP/p300 rescued Smad-specific gene transcription in the presence of cAMP suggesting that sequestration of limited amounts of CBP/p300 by the activated cAMP/CREB pathway is the molecular basis of this inhibitory effect. These findings were extended by two functional assays. Increased intracellular cAMP levels suppressed the inductive activity of TGF-beta to contract mechanically unloaded collagen lattices and resulted in an attenuation of fibroblast migration of mechanically induced cell layer wounds. Of note, cAMP and TGF-beta synergistically induced hyaluronan synthase 2 (HAS2) expression and hyaluronan secretion, presumably via putative CREB-binding sites adjacent to Smad-binding sites within the HAS2 promoter. Our findings identify the cAMP pathway as a potent but differential and promoter-specific regulator of TGF-beta-mediated effects involved in extracellular matrix homeostasis.
Collapse
Affiliation(s)
- Meinhard Schiller
- Department of Dermatology, Ludwig Boltzmann Institute for Cell Biology and Immunobiology of the Skin, 48149 Münster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Loss of steroid receptor co-activator-3 attenuates carbon tetrachloride-induced murine hepatic injury and fibrosis. J Transl Med 2009; 89:903-14. [PMID: 19488034 PMCID: PMC3620314 DOI: 10.1038/labinvest.2009.51] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hepatic fibrosis, a disease characterized by altered accumulation of extracellular matrix, can cause cirrhosis and liver failure. There is growing interest in the impact of co-activators on hepatic fibrogenesis. Here, we provided genetic evidence that mice lacking steroid receptor co-activator-3 (SRC-3) were protected against carbon tetrachloride (CCl4)-induced acute liver necrosis and chronic hepatic fibrosis. After acute CCl4 treatment, SRC-3(-/-) mice showed attenuated profibrotic response and hepatocyte apoptosis, whereas hepatocyte proliferation was elevated in SRC-3(-/-) mice versus SRC-3+/+ mice. Similarly, chronically CCl4-treated SRC-3(-/-) mice showed significant weakening of inflammatory infiltrates, hepatic stellate cell activation and collagen accumulation in the liver compared with SRC-3+/+ mice. Further investigation revealed that TGFbeta1/Smad signaling pathway was impaired in the absence of SRC-3. Moreover, the expression levels of SRC-3, as assessed in human tissue microarray of liver diseases, correlated positively with degrees of fibrosis. These data revealed that SRC-3(-/-) mice were resistant to CCl4-induced acute and chronic hepatic damage and TGFbeta1/Smad signaling was suppressed in the lack of SRC-3. Our results established an essential involvement of SRC-3 in liver fibrogenesis, which might provide new clues to the future treatment of hepatic fibrosis.
Collapse
|
13
|
Loss of steroid receptor co-activator-3 attenuates carbon tetrachloride-induced murine hepatic injury and fibrosis. LABORATORY INVESTIGATION; A JOURNAL OF TECHNICAL METHODS AND PATHOLOGY 2009. [PMID: 19488034 DOI: 10.1038/labinvest] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatic fibrosis, a disease characterized by altered accumulation of extracellular matrix, can cause cirrhosis and liver failure. There is growing interest in the impact of co-activators on hepatic fibrogenesis. Here, we provided genetic evidence that mice lacking steroid receptor co-activator-3 (SRC-3) were protected against carbon tetrachloride (CCl4)-induced acute liver necrosis and chronic hepatic fibrosis. After acute CCl4 treatment, SRC-3(-/-) mice showed attenuated profibrotic response and hepatocyte apoptosis, whereas hepatocyte proliferation was elevated in SRC-3(-/-) mice versus SRC-3+/+ mice. Similarly, chronically CCl4-treated SRC-3(-/-) mice showed significant weakening of inflammatory infiltrates, hepatic stellate cell activation and collagen accumulation in the liver compared with SRC-3+/+ mice. Further investigation revealed that TGFbeta1/Smad signaling pathway was impaired in the absence of SRC-3. Moreover, the expression levels of SRC-3, as assessed in human tissue microarray of liver diseases, correlated positively with degrees of fibrosis. These data revealed that SRC-3(-/-) mice were resistant to CCl4-induced acute and chronic hepatic damage and TGFbeta1/Smad signaling was suppressed in the lack of SRC-3. Our results established an essential involvement of SRC-3 in liver fibrogenesis, which might provide new clues to the future treatment of hepatic fibrosis.
Collapse
|
14
|
Mishra R, Cool BL, Laderoute KR, Foretz M, Viollet B, Simonson MS. AMP-activated protein kinase inhibits transforming growth factor-beta-induced Smad3-dependent transcription and myofibroblast transdifferentiation. J Biol Chem 2008; 283:10461-9. [PMID: 18250161 DOI: 10.1074/jbc.m800902200] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In wound healing, myofibroblast transdifferentiation (MFT) is a metaplastic change in phenotype producing profibrotic effector cells that secrete and remodel the extracellular matrix. Unlike pathways that induce MFT, the molecular mechanisms that negatively regulate MFT are poorly understood. Here, we report that AMP-activated protein kinase (AMPK) blocks MFT in response to transforming growth factor-beta (TGFbeta). Pharmacological activation of AMPK inhibited TGFbeta-induced secretion of extracellular matrix proteins collagen types I and IV and fibronectin. AMPK activation also prevented induction of the myofibroblast phenotype markers alpha-smooth muscle actin and the ED-A fibronectin splice variant. AMPK activators did not prevent MFT in cells transduced with an adenovirus expressing dominant negative, kinase-dead AMPKalpha2. Moreover, AMPK activators did not inhibit MFT induction in AMPK(alpha1,2)(-/-) fibroblasts, demonstrating a requirement for AMPK(alpha) expression. Adenoviral transduction of constitutively active AMPK(alpha2) was sufficient to prevent TGFbeta-induced collagen I, alpha-smooth muscle actin, and ED-A fibronectin. AMPK did not reduce TGFbeta-stimulated Smad3 COOH-terminal phosphorylation and nuclear translocation, which are necessary for MFT. However, AMPK activation inhibited TGFbeta-induced transcription driven by Smad3-binding cis-elements. Consistent with a role for AMPK in transcriptional regulation, nuclear translocation of AMPKalpha2 correlated with the appearance of active AMPKalpha in the nucleus. Collectively, these results demonstrate that AMPK inhibits TGFbeta-induced transcription downstream of Smad3 COOH-terminal phosphorylation and nuclear translocation. Furthermore, activation of AMPK is sufficient to negatively regulate MFT in vitro.
Collapse
Affiliation(s)
- Rangnath Mishra
- Division of Nephrology and Hypertension, Department of Medicine, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
| | | | | | | | | | | |
Collapse
|
15
|
Ross S, Hill CS. How the Smads regulate transcription. Int J Biochem Cell Biol 2007; 40:383-408. [PMID: 18061509 DOI: 10.1016/j.biocel.2007.09.006] [Citation(s) in RCA: 289] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 09/19/2007] [Accepted: 09/20/2007] [Indexed: 01/01/2023]
Abstract
The primary signalling pathway downstream of ligands of the transforming growth factor beta (TGF-beta) superfamily is the Smad pathway. Activated receptors phosphorylate receptor-regulated Smads, which form homomeric complexes and heteromeric complexes with Smad4. These activated Smad complexes accumulate in the nucleus, where they are directly involved in the regulation of transcription of target genes. This apparently very simple pathway is subject to complex regulation, much of which is at the level of post-translational modifications of pathway components, in particular, the Smads. The enzymes responsible may be constitutively active, may be cell type-specific or may be regulated by other signalling pathways or by the cell cycle. In this way, signals from TGF-beta superfamily ligands are integrated with signals from other growth factors and cytokines, are regulated by the cell cycle and are dependent on cell type. This may go some way to explaining the pleiotropic nature of TGF-beta superfamily responses. In this review we focus on the mechanisms whereby the Smads are modified and regulated. We then go on to discuss how the activated Smad complexes regulate transcription.
Collapse
Affiliation(s)
- Sarah Ross
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, Lincoln's Inn Fields Laboratories, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | | |
Collapse
|
16
|
Cherlet T, Murphy LC. Estrogen receptors inhibit Smad3 transcriptional activity through Ap-1 transcription factors. Mol Cell Biochem 2007; 306:33-42. [PMID: 17660955 DOI: 10.1007/s11010-007-9551-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Accepted: 07/12/2007] [Indexed: 02/07/2023]
Abstract
Breast tumorigenesis and breast cancer progression involves the deregulation or hyperactivation of intracellular signaling proteins that leads to uncontrolled cellular proliferation, invasion and metastasis. For example, the expression and cellular responses to estogen receptor (ER) and transforming growth factor beta (TGFbeta) signaling pathways change during breast tumorigenesis and breast cancer progression. While the expression and activity of ER and TGFbeta maybe significant in the development of breast cancer, alterations in the cross-talk between these pathways may be equally important. Autocrine and paracrine effects of TGFbeta on breast cancer cell growth have been known for some time, but only recently have direct interactions between ER and TGFbeta been described. The purpose of this article was to further characterize the cross-talk between ER and TGFbeta, by examining ER interaction with Smad3, a downstream mediator of TGFbeta signaling. Transient transfection of Cos1 cells with p3TP-lux, demonstrate that ERalpha and ERbeta(1) repress Smad3 transcriptional activity in an estradiol-dependent manner and that this effect is inhibited by antiestrogen treatment. The ERbeta variants, ERbeta(2) and ERbeta(5), did not have any effect on Smad3 transcriptional activity. Further experiments attempted to characterize the molecular mechanism by which activated ER inhibits Smad3 transcriptional activity. Results indicate that ligand-bound ER does not affect Smad3 protein expression levels and that ER does not form direct protein interactions with Smad3. Transient transfection of Cos1 cells with the Ap-1 transcription factor c-Jun but not c-Fos was able to rescue the inhibitory effect of estrogen on Smad3 transcriptional activity. Based on these results, a model is proposed whereby c-Jun is limiting in its ability to act as a Smad3 co-activator in the presence of E(2)-bound ER, possibly due to ER sequestering c-Jun away from the Smad3 responsive promoter.
Collapse
Affiliation(s)
- Tracy Cherlet
- Manitoba Institute of Cell Biology, Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB, Canada R3E 0V9
| | | |
Collapse
|
17
|
Abstract
Transforming growth factor-β (TGF-β), a prototype of multifunctional cytokine, is a key regulator of extracellular matrix (ECM) assembly and remodeling. Specifically, TGF-β isoforms have the ability to induce the expression of ECM proteins in mesenchymal cells, and to stimulate the production of protease inhibitors that prevent enzymatic breakdown of the ECM. Elevated TGF-β expression in affected organs, and subsequent deregulation of TGF-β functions, correlates with the abnormal connective tissue deposition observed during the onset of fibrotic diseases. During the last few years, tremendous progress has been made in the understanding of the molecular aspects of intracellular signaling downstream of the TGF-β receptors. In particular, Smad proteins, TGF-β receptor kinase substrates that translocate into the cell nucleus to act as transcription factors, have been studied extensively. The role of Smad3 in the transcriptional regulation of typeIcollagen gene expression and in the development of fibrosis, demonstrated both in vitro and in animal models with a targeted deletion of Smad3, is of critical importance because it may lead to novel therapeutic strategies against these diseases. This review focuses on the mechanisms underlying Smad modulation of fibrillar collagen expression and how it relates to fibrotic processes.
Collapse
Affiliation(s)
- Franck Verrecchia
- INSERM U697, Hopital Saint-Louis, Pavillon Bazin, 1 avenue Claude Vellefaux, Paris 75010, France.
| | | |
Collapse
|
18
|
Chou YT, Wang H, Chen Y, Danielpour D, Yang YC. Cited2 modulates TGF-beta-mediated upregulation of MMP9. Oncogene 2006; 25:5547-60. [PMID: 16619037 DOI: 10.1038/sj.onc.1209552] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cited (CBP/p300-interacting transactivators with glutamic acid (E)/aspartic acid (D)-rich C-terminal domain) 2, which is a CBP/p300-binding transcription co-activator without typical DNA-binding domains, has been implicated in control of cell growth and malignant transformation in Rat1 cells. In this report, we provide evidence that Cited2 is an important regulator of transforming growth factor (TGF)-beta signaling. Overexpression of Cited2 enhanced TGF-beta-mediated transcription of a Smad-Binding Element-containing luciferase reporter construct, SBE4-Luc. This may occur through a direct physical association of Cited2 with Smads 2 and 3, as supported by co-immunoprecipitation, mammalian two-hybrid and glutathione S-transferase-pull down assays. The transcription factor p300, which binds to Smad3, was shown to further enhance the interaction between Cited2 and Smad3, and the transcriptional responses of Smad3 by Cited2 in reporter assays. Cited2 enhances TGF-beta-mediated upregulation of matrix metalloproteinase 9 (MMP9) in Cited2 inducible mouse embryo fibroblasts. Overexpression of Cited2 enhanced TGF-beta-mediated MMP9 promoter reporter activity. Moreover, knockdown of Cited2 in MDA-MB-231 cells attenuated TGF-beta-mediated upregulation of MMP9 and TGF-beta-mediated cell invasion. Chromatin immunoprecipitation showed that Cited2 and Smad3 were recruited to MMP9 promoter upon TGF-beta stimulation. This is the first demonstration that Cited2 functions as a Smad3/p300-interacting transcriptional co-activator in modulating the expression of MMP9, which could affect tumor cell invasion mediated by TGF-beta.
Collapse
Affiliation(s)
- Y-T Chou
- Department of Pharmacology and Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106-4965, USA
| | | | | | | | | |
Collapse
|