1
|
Bai Y, Liu F, Luo S, Wan Y, Zhang L, Wu X, Chen Q, Xie Y, Guo P. Experimental study on H 2O 2 activation of HSC-T6 and hepatic fibrosis in cholestatic mice by "Yajieshaba". JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118712. [PMID: 39173724 DOI: 10.1016/j.jep.2024.118712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 08/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yajieshaba (YJSB), approved by the Yunnan Provincial Food and Drug Administration in 2008, are known for their anti-inflammatory, antiviral, and pro-apoptotic properties, effectively treating Hepatic fibrosis (HF). However, its mechanism of action remains unclear. AIM OF THE STUDY The objective of this investigation is to explore how YJSB influences the TGF-β1/Smad signaling pathway as a strategy for reducing HF. METHODS The establishment of a HF model in mice involved ligation of the common bile duct, followed by administration of YJSB. Body and liver weights were measured, and the liver index calculated. Serum levels of ALT, AST, ALP, TBA, and TBIL were assessed using colorimetric methods. Additionally, liver homogenates were analyzed for PIIINP, Col-IV, LN, HA, and Hyp, as well as TGF-β1 activity, using ELISA. Histological analyses of liver sections, stained with H&E, Ag, and Masson's trichrome, were performed to examine inflammation and the accumulation of collagen and reticular fibers. These studies aimed to elucidate the pharmacodynamic effects of YJSB on HF in mice with bile duct obstruction. The target pathways of YJSB were preliminarily identified through immunofluorescence detection of TGF-β1, P-Smad2L, P-Smad2C, P-Smad3L, P-Smad3C, and Smad4 proteins. In vitro experiments included the induction of hepatic stellate cell (HSC-T6) activation by H2O2. A cell injury model was established for HSC-T6, and the CCK-8 assay was used to determine the optimal YJSB concentration and treatment duration. After pirfenidone (PFD) administration, which inhibits the TGF-β1/Smad pathway, the effects of YJSB on HSC-T6 cell proliferation were observed. ELISA assays quantified Col-III, α-SMA, and Col-I in cell lysates to assess YJSB's impact on collagen synthesis in HSC-T6 cells. Western blot analysis was performed to assess the protein levels within the TGF-β1/Smad signaling cascade. RESULTS In the HF mouse model, administration of YJSB notably augmented the body weight and reduced the liver index. Concurrently, there was an elevation in serum concentrations of ALP, AST, ALT, TBA, and TBIL. Similarly, in the liver homogenates of HF mice, increases were observed in the levels of HA, PIIINP, Col-IV, LN, Hyp, and TGF-β1. Histological assessments using H&E, Ag, and Masson stains indicated a substantial diminution in liver tissue damage. Through immunofluorescence analysis, it was discerned that YJSB modulated the expression of TGF-β1, P-Smad2L, P-Smad2C, and P-Smad3L downwards, while elevating P-Smad3C and Smad4 protein expressions. Additional investigations revealed a significant reduction in α-SMA, Col-I, and Col-III levels in cell culture fluids, suggesting a decrease in collagen synthesis and a protective role against cellular damage. Western blot analyses demonstrated that the TGF-β1/Smad pathway inhibitor, PFD, acted in synergy with YJSB, enhancing its regulatory effects on this pathway, decreasing levels of TGF-β1, P-Smad2L, P-Smad2C, P-Smad3L, and promoting the expression of P-Smad3C. CONCLUSIONS YJSB demonstrates a pharmacodynamic effect against HF, enhancing liver functionality and effectively mitigating the damage associated with bile duct obstruction. The proposed action mechanism of YJSB involves modulation of the TGF-β1/Smad signaling pathway. Research indicates that YJSB might play a role in suppressing the movement, programmed cell death, and activation of HSC-T6, potentially decelerating the advancement of hepatic fibrosis.
Collapse
Affiliation(s)
- Yuanmei Bai
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Yunnan, Kunming, 650500, China
| | - Feifan Liu
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Yunnan, Kunming, 650500, China
| | - Shifang Luo
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Yunnan, Kunming, 650500, China
| | - Yan Wan
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Yunnan, Kunming, 650500, China
| | - Linao Zhang
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Yunnan, Kunming, 650500, China
| | - Xue Wu
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Yunnan, Kunming, 650500, China
| | - Qinghua Chen
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Yunnan, Kunming, 650500, China.
| | - Yuhuan Xie
- College of Basic Medical Sciences, Yunnan University of Chinese Medicine, Yunnan, Kunming, 650500, China.
| | - Peixin Guo
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Yunnan, Kunming, 650500, China.
| |
Collapse
|
2
|
Peng S, Hu L, Ge W, Deng J, Yao L, Li H, Xu D, Mo H. ChIP-Seq Analysis of AtfA Interactions in Aspergillus flavus Reveals Its Involvement in Aflatoxin Metabolism and Virulence Under Oxidative Stress. Int J Mol Sci 2024; 25:12213. [PMID: 39596279 PMCID: PMC11594458 DOI: 10.3390/ijms252212213] [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: 10/29/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
The risk of Aspergillus flavus contamination is expanding with global warming. Targeting the pathogenicity of A. flavus at its source and diminishing its colonization within the host may be a potential control strategy. Oxidative stress transcription factor AtfA plays a pivotal role in A. flavus pathogenicity by combating reactive oxygen species (ROS) generated by host immune cells. This study employed chromatin immunoprecipitation sequencing to elucidate the binding sites and epigenetic mechanisms of AtfA under oxidative stress. Among the total 1022 identified potential AtfA-binding peaks, a 10-bp region predominated by 5'-DRTGTTGCAA-3', which is highly similar to the AP-1 binding motif was predicted. The significantly regulated genes exhibited a variety of biological functions, including regulation of filamentous growth, response to extracellular stimulus, and regulation of gene expression. Moreover, AtfA indirectly influenced these processes via the MAPK signaling pathway, carbon metabolism, and fatty acid metabolism in response to oxidative stress. The absence of atfA contributed to the decrease in the growth and development, sporulation, AFB1 biosynthesis, and invasion ability of A. flavus under oxidative stress. These findings suggest that AtfA is critical to overcome oxidative stress induced by the host immune cells during the infection, providing a novel target for early prevention of A. flavus contamination.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dan Xu
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (S.P.); (L.H.); (W.G.); (J.D.); (L.Y.); (H.L.)
| | - Haizhen Mo
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (S.P.); (L.H.); (W.G.); (J.D.); (L.Y.); (H.L.)
| |
Collapse
|
3
|
Lin Z, Wang Y, Deng Y, Li L, Cao Y, Wang S, Zhang X, Ding G, Cheng J, Tang S, Zhou J. Jun modulates endoplasmic reticulum stress-associated ferroptosis in dorsal root ganglia neurons during neuropathic pain by regulating Timp1. Neurochem Int 2024; 180:105877. [PMID: 39384071 DOI: 10.1016/j.neuint.2024.105877] [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: 08/06/2024] [Revised: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 10/11/2024]
Abstract
Neuropathic pain (NP) is a complex disorder caused by lesions or diseases affecting the somatosensory nervous system, severely impacting patients' quality of life. Recent studies suggest ferroptosis may be involved in NP induction, but its precise mechanisms remain unclear. We used GO and KEGG pathway enrichment analyses to functionally annotate ferroptosis-related differentially expressed genes (FRDs). Through STRING and the maximum cluster centrality (MCC) algorithm, we identified five hub FRDs (Jun, Timp1, Egfr, Cdkn1a, Cdkn2a). Single-cell analysis revealed significant expression of Jun and Timp1 in neurons. Our study confirmed the association between ferroptosis and endoplasmic reticulum stress (ERS) in NP and validated changes in hub FRD expression across various NP animal models. In vitro experiments demonstrated that Jun regulates neuronal ferroptosis and ERS, particularly by modulating Timp1 expression. Transcription factor prediction and JASPAR binding site analysis elucidated the regulatory network involving Jun. ROC curve analysis of external datasets highlighted the diagnostic potential of hub FRDs and ERS-related differentially expressed genes (ERSRDs) in NP. Using the Comparative Toxicogenomics Database (CTD), we identified estradiol (E2) as a potential therapeutic drug targeting hub FRDs and ERSRDs. Molecular docking predicted its binding sites with Jun and Timp1, and in vivo experiments confirmed that E2 alleviated NP and reversed the expression of Jun and Timp1. This study underscores the crucial role of Jun and Timp1 in the interplay between ferroptosis and ERS, offering new insights and promising avenues for NP treatment.
Collapse
Affiliation(s)
- Ziqiang Lin
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Yi Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Yingdong Deng
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Lu Li
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Yu Cao
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Suo Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, China
| | - Xiangsheng Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Guoda Ding
- Postgraduate Training Base of Hubei University of Medicine, Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Jiurong Cheng
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Simin Tang
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Jun Zhou
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510000, Guangdong, China.
| |
Collapse
|
4
|
Ciceri S, Bertolotti A, Serra A, Gattuso G, Boschetti L, Capasso M, Cecchi C, Sorrentino S, Quarello P, Ciniselli CM, Verderio P, De Cecco L, Manenti G, Diomedi Camassei F, Collini P, Spreafico F, Perotti D. Widening the spectrum of players affected by genetic changes in Wilms tumor relapse. iScience 2024; 27:110684. [PMID: 39262773 PMCID: PMC11387809 DOI: 10.1016/j.isci.2024.110684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024] Open
Abstract
Few studies investigated the genetics of relapsed Wilms tumor (WT), suggesting the SIX1 gene, the microRNA processing genes, and the MYCN network as possibly involved in a relevant percentage of relapses. We investigated 28 relapsing WT patients (10 new cases and 18 cases in which the involvement of SIX and miRNAPG had been excluded) with a panel of ∼5000 genes. We identified variants affecting genes involved in DNA damage prevention and repair in 12/28 relapsing patients (42.9%), and affecting genes involved in chromatin modification and regulation in 6/28 relapsing patients (21.4%), widening the spectrum of anomalies detected in relapsed tumors. The disclosure of molecular pathways possibly underlying tumor progression might allow to use molecularly targeted therapies at relapse. Surprisingly, germline anomalies, mostly affecting DNA damage prevention and repair genes, were identified in 13/28 patients (46.4%), raising the issue of performing a genetic testing to all children presenting with a WT.
Collapse
Affiliation(s)
- Sara Ciceri
- Predictive Medicine: Molecular Bases of Genetic Risk, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Alessia Bertolotti
- Diagnostic and Molecular Research Lab, Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Annalisa Serra
- Department of Pediatric Hematology and Oncology, Gene and Cellular Therapy, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Giovanna Gattuso
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Luna Boschetti
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Maria Capasso
- Department of Pediatric Hemato-Oncology, AORN Santobono-Pausilipon, Naples, Italy
| | - Cecilia Cecchi
- Division of Pediatric Oncology/Hematology, Meyer University Children's Hospital, Florence, Italy
| | | | - Paola Quarello
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Turin, Italy
- Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| | - Chiara Maura Ciniselli
- Unit of Bioinformatics and Biostatistics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Paolo Verderio
- Unit of Bioinformatics and Biostatistics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Loris De Cecco
- Integrated Biology of Rare Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Giacomo Manenti
- Unit of Animal Health and Welfare, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | | | - Paola Collini
- Soft Tissue Tumor Pathology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Filippo Spreafico
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Daniela Perotti
- Predictive Medicine: Molecular Bases of Genetic Risk, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
5
|
Zelisko N, Lesyk R, Stoika R. Structure, unique biological properties, and mechanisms of action of transforming growth factor β. Bioorg Chem 2024; 150:107611. [PMID: 38964148 DOI: 10.1016/j.bioorg.2024.107611] [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: 01/21/2024] [Revised: 06/07/2024] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
Transforming growth factor β (TGF-β) is a ubiquitous molecule that is extremely conserved structurally and plays a systemic role in human organism. TGF-β is a homodimeric molecule consisting of two subunits joined through a disulphide bond. In mammals, three genes code for TGF-β1, TGF-β2, and TGF-β3 isoforms of this cytokine with a dominating expression of TGF-β1. Virtually, all normal cells contain TGF-β and its specific receptors. Considering the exceptional role of fine balance played by the TGF-β in anumber of physiological and pathological processes in human body, this cytokine may be proposed for use in medicine as an immunosuppressant in transplantology, wound healing and bone repair. TGFb itself is an important target in oncology. Strategies for blocking members of TGF-β signaling pathway as therapeutic targets have been considered. In this review, signalling mechanisms of TGF-β1 action are addressed, and their role in physiology and pathology with main focus on carcinogenesis are described.
Collapse
Affiliation(s)
- Nataliya Zelisko
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine.
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine
| |
Collapse
|
6
|
Zhang Z, Li B, Wu S, Yang Y, Wu B, Lai Q, Lai F, Mo F, Zhong Y, Wang S, Guo R, Zhang B. Bergenin protects against osteoarthritis by inhibiting STAT3, NF-κB and Jun pathways and suppressing osteoclastogenesis. Sci Rep 2024; 14:20292. [PMID: 39217193 PMCID: PMC11366014 DOI: 10.1038/s41598-024-71259-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease characterized by articular cartilage destruction and subchondral bone reconstruction in the early stages. Bergenin (Ber) is a cytoprotective polyphenol found in many medicinal plants. It has been proven to have anti-inflammatory, antioxidant, and other biological activities, which may reveal its potential role in the treatment of OA. This study aimed to determine the potential efficacy of Ber in treating OA and explore the possible underlying mechanism through network pharmacology and validation experiments. The potential co-targets and processes of Ber and OA were predicted by using network pharmacology, including a Venn diagram for intersection targets, a protein‒protein interaction (PPI) network to obtain key potential targets, and GO and KEGG pathway enrichment to reveal the probable mechanism of action of Ber on OA. Subsequently, validation experiments were carried out to investigate the effects and mechanisms of Ber in treating OA in vitro and vivo. Ber suppressed IL-1β-induced chondrocyte apoptosis and extracellular matrix catabolism by inhibiting the STAT3, NF-κB and Jun signalling pathway in vitro. Furthermore, Ber suppressed the expression of osteoclast marker genes and RANKL-induced osteoclastogenesis. Ber alleviated the progression of OA in DMM-induced OA mice model. These results demonstrated the protective efficacy and potential mechanisms of Ber against OA, which suggested that Ber could be adopted as a potential therapeutic agent for treating OA.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Bo Li
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Shuqin Wu
- Faculty of Jiangxi Medical College, Donghu District, Nanchang University, No.461 Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Yuxin Yang
- Faculty of Jiangxi Medical College, Donghu District, Nanchang University, No.461 Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Binkang Wu
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Qi Lai
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Fuchong Lai
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Fengbo Mo
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Yufei Zhong
- Faculty of Jiangxi Medical College, Donghu District, Nanchang University, No.461 Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Song Wang
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
| | - Runsheng Guo
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
| | - Bin Zhang
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
7
|
Liu X, Ping G, Ji D, Wen Z, Chen Y. Reclassify High-Grade Serous Ovarian Cancer Patients Into Different Molecular Subtypes With Discrepancy Prognoses and Therapeutic Responses Based on Cancer-Associated Fibroblast-Enriched Prognostic Genes. Biomed Eng Comput Biol 2024; 15:11795972241274024. [PMID: 39221174 PMCID: PMC11365035 DOI: 10.1177/11795972241274024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) play critical roles in the metastasis and therapeutic response of high-grade serous ovarian cancer (HGSC). Our study intended to select HGSC patients with unfavorable prognoses and therapeutic responses based on CAF-enriched prognostic genes. The bulk RNA and single-cell RNA sequencing (scRNA-seq) data of tumor tissues were collected from the TCGA and GEO databases. The infiltrated levels of immune and stromal cells were estimated by multiple immune deconvolution algorithms and verified through immunohistochemical analysis. The univariate Cox regression analyses were used to identify prognostic genes. Gene Set Enrichment Analysis (GSEA) was conducted to annotate enriched gene sets. The Genomics of Drug Sensitivity in Cancer (GDSC) database was used to explore potential alternative drugs. We found the infiltered levels of CAFs were remarkedly elevated in advanced and metastatic HGSC tissues and identified hundreds of genes specifically enriched in CAFs. Then we selected 6 CAF-enriched prognostic genes based on which HGSC patients were reclassified into 2 subclusters with discrepancy prognoses. Further analysis revealed that the HGSC patients in cluster-2 tended to undergo poor responses to traditional chemotherapy and immunotherapy. Subsequently, we selected 24 novel potential therapeutic drugs for cluster-2 HGSC patients. Moreover, we discovered a positive correlation of infiltrated levels between CAFs and monocytes/macrophages in HGSC tissues. Collectively, our study successfully reclassified HGSC patients into 2 different subgroups that have discrepancy prognoses and responses to current therapeutic methods.
Collapse
Affiliation(s)
- Xiangxiang Liu
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, Jiangsu, China
| | - Guoqiang Ping
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongze Ji
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhifa Wen
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, Jiangsu, China
| | - Yajun Chen
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Hasselluhn MC, Schlösser D, Versemann L, Schmidt GE, Ulisse M, Oschwald J, Zhang Z, Hamdan F, Xiao H, Kopp W, Spitalieri J, Kellner C, Schneider C, Reutlinger K, Nagarajan S, Steuber B, Sastra SA, Palermo CF, Appelhans J, Bohnenberger H, Todorovic J, Kostyuchek I, Ströbel P, Bockelmann A, König A, Ammer-Herrmenau C, Schmidleitner L, Kaulfuß S, Wollnik B, Hahn SA, Neesse A, Singh SK, Bastians H, Reichert M, Sax U, Olive KP, Johnsen SA, Schneider G, Ellenrieder V, Hessmann E. An NFATc1/SMAD3/cJUN Complex Restricted to SMAD4-Deficient Pancreatic Cancer Guides Rational Therapies. Gastroenterology 2024; 166:298-312.e14. [PMID: 37913894 DOI: 10.1053/j.gastro.2023.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 09/19/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND & AIMS The highly heterogeneous cellular and molecular makeup of pancreatic ductal adenocarcinoma (PDAC) not only fosters exceptionally aggressive tumor biology, but contradicts the current concept of one-size-fits-all therapeutic strategies to combat PDAC. Therefore, we aimed to exploit the tumor biological implication and therapeutic vulnerabilities of a clinically relevant molecular PDAC subgroup characterized by SMAD4 deficiency and high expression of the nuclear factor of activated T cells (SMAD4-/-/NFATc1High). METHODS Transcriptomic and clinical data were analyzed to determine the prognostic relevance of SMAD4-/-/NFATc1High cancers. In vitro and in vivo oncogenic transcription factor complex formation was studied by immunoprecipitation, proximity ligation assays, and validated cross model and species. The impact of SMAD4 status on therapeutically targeting canonical KRAS signaling was mechanistically deciphered and corroborated by genome-wide gene expression analysis and genetic perturbation experiments, respectively. Validation of a novel tailored therapeutic option was conducted in patient-derived organoids and cells and transgenic as well as orthotopic PDAC models. RESULTS Our findings determined the tumor biology of an aggressive and chemotherapy-resistant SMAD4-/-/NFATc1High subgroup. Mechanistically, we identify SMAD4 deficiency as a molecular prerequisite for the formation of an oncogenic NFATc1/SMAD3/cJUN transcription factor complex, which drives the expression of RRM1/2. RRM1/2 replenishes nucleoside pools that directly compete with metabolized gemcitabine for DNA strand incorporation. Disassembly of the NFATc1/SMAD3/cJUN complex by mitogen-activated protein kinase signaling inhibition normalizes RRM1/2 expression and synergizes with gemcitabine treatment in vivo to reduce the proliferative index. CONCLUSIONS Our results suggest that PDAC characterized by SMAD4 deficiency and oncogenic NFATc1/SMAD3/cJUN complex formation exposes sensitivity to a mitogen-activated protein kinase signaling inhibition and gemcitabine combination therapy.
Collapse
Affiliation(s)
- Marie C Hasselluhn
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Department of Medicine, Division of Digestive and Liver Diseases, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Denise Schlösser
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany
| | - Lennart Versemann
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany
| | - Geske E Schmidt
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany
| | - Maria Ulisse
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany
| | - Joana Oschwald
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany
| | - Zhe Zhang
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany
| | - Feda Hamdan
- Gene Regulatory Mechanisms and Molecular Epigenetics Laboratory, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Harry Xiao
- Department of Medicine, Division of Digestive and Liver Diseases, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Waltraut Kopp
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany
| | - Jessica Spitalieri
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany
| | - Christin Kellner
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany
| | - Carolin Schneider
- Department of General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Goettingen, Germany
| | - Kristina Reutlinger
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany
| | - Sankari Nagarajan
- Manchester Breast Centre and Manchester Cancer Research Centre, Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Benjamin Steuber
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany
| | - Stephen A Sastra
- Department of Medicine, Division of Digestive and Liver Diseases, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Carmine F Palermo
- Department of Medicine, Division of Digestive and Liver Diseases, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Jennifer Appelhans
- Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany; Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Jovan Todorovic
- Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany; Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Irina Kostyuchek
- Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Philipp Ströbel
- Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany; Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Aiko Bockelmann
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany
| | - Alexander König
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany
| | - Christoph Ammer-Herrmenau
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany
| | - Laura Schmidleitner
- Medical Clinic and Polyclinic II, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany; Translational Pancreatic Research Cancer Center, Medical Clinic and Polyclinic II, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Silke Kaulfuß
- Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany; Institute of Human Genetics, University Medical Center Goettingen, Goettingen, Germany
| | - Bernd Wollnik
- Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany; Institute of Human Genetics, University Medical Center Goettingen, Goettingen, Germany; Cluster of Excellence Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells, University of Goettingen, Germany
| | - Stephan A Hahn
- Ruhr University Bochum, Faculty of Medicine, Department of Molecular Gastrointestinal Oncology, Bochum, Germany
| | - Albrecht Neesse
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany
| | - Shiv K Singh
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany
| | - Holger Bastians
- Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany; Department of Molecular Oncology, Section for Cellular Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Maximilian Reichert
- Medical Clinic and Polyclinic II, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany; Translational Pancreatic Research Cancer Center, Medical Clinic and Polyclinic II, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany; German Cancer Consortium (a partnership between Deutsches Krebsforschungszentrum and University Hospital Klinikum Rechts der Isar), Munich, Germany; Center for Protein Assemblies, Technical University of Munich, Garching, Germany; Center for Organoid Systems and Tissue Engineering, Technical University Munich, Garching, Germany
| | - Ulrich Sax
- Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany; Department of Medical Informatics, University Medical Center Goettingen, Goettingen, Germany
| | - Kenneth P Olive
- Department of Medicine, Division of Digestive and Liver Diseases, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Steven A Johnsen
- Department of General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Goettingen, Germany; Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Günter Schneider
- Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany; Department of General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Goettingen, Germany; Comprehensive Cancer Center, Lower Saxony, Goettingen and Hannover, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany; Comprehensive Cancer Center, Lower Saxony, Goettingen and Hannover, Germany
| | - Elisabeth Hessmann
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany; Clinical Research Unit KFO5002, University Medical Center Goettingen, Goettingen, Germany; Comprehensive Cancer Center, Lower Saxony, Goettingen and Hannover, Germany.
| |
Collapse
|
9
|
Zhang H, Liao Z, Wang W, Liu Y, Zhu H, Liang H, Zhang B, Chen X. A micropeptide JunBP regulated by TGF-β promotes hepatocellular carcinoma metastasis. Oncogene 2023; 42:113-123. [PMID: 36380240 PMCID: PMC9816058 DOI: 10.1038/s41388-022-02518-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022]
Abstract
Transforming growth factor beta (TGF-β) signaling pathway plays important roles in hepatocellular carcinoma (HCC) progression. Long intergenic non-protein coding RNAs (lincRNAs) are important components of TGF-β signaling pathway and perform their functions through different mechanisms. Here, we found that LINC02551 was activated by TGF-β transcriptionally and identified a 174-amino-acid peptide, Jun binding micropeptide (JunBP), encoded by LINC02551 in HCC tissues and HCC cell lines. Functional study showed that JunBP promotes HCC metastasis through binding to c-Jun and subsequent promotion of its phosphorylated activation. Activated c-Jun has higher binding affinity to SMAD3, which in turn leads to more SMAD3 recruited to the promoter region of LINC02551. We find a positive feedback among them, and this mechanism provides a novel potential prognostic biomarker and therapeutic target in HCC.
Collapse
Affiliation(s)
- Hongwei Zhang
- grid.33199.310000 0004 0368 7223Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei P.R. China ,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei P.R. China
| | - Zhibin Liao
- grid.33199.310000 0004 0368 7223Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei P.R. China ,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei P.R. China
| | - Weijian Wang
- grid.33199.310000 0004 0368 7223Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei P.R. China ,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei P.R. China
| | - Yachong Liu
- grid.33199.310000 0004 0368 7223Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei P.R. China ,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei P.R. China
| | - He Zhu
- grid.33199.310000 0004 0368 7223Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei P.R. China ,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei P.R. China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China. .,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, P.R. China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China. .,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, P.R. China. .,Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Wuhan, Hubei, P.R. China.
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China. .,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, P.R. China. .,Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Wuhan, Hubei, P.R. China.
| |
Collapse
|
10
|
Cai X, Zha H, Yang Z, Du Y, Dai X, Yang B, Wang J, He Q, Weng Q. Genetic dominance of transforming growth factor-β1 polymorphisms in chronic liver disease. Front Immunol 2022; 13:1058532. [DOI: 10.3389/fimmu.2022.1058532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic liver disease (CLD) is an extremely common clinical condition accompanied by sustained inflammatory response leading to tissue damage. Transforming growth factor-β1 (TGF-β1) is known as a master immune regulator in CLDs, but the association between TGF-β1 polymorphisms and CLD risk is controversial and inconclusive, and the genetic dominance of CLDs remains unknown. In this study, the relationship between TGF-β1 polymorphisms and CLD susceptibility is systematically analyzed based on 35 eligible studies. Individuals with the TGF-β1-509 allele (TT or CT) or codon 10 allele (Pro/Pro) show an increased risk of CLDs. Subgroup analyses indicate TGF-β1-509C/T has a significant correlation with cirrhosis and chronic hepatitis C, codon 10 is associated with chronic hepatitis B occurrence, and codon 25 exhibits a relationship with autoimmune hepatitis risk. Missense mutations in G29E, A105S, D191N, and F321L of TGF-β1 are the genetic factors of HCC susceptibility. Furthermore, the TGF-β1 gene expression is significantly elevated in CLD patients, and the TGF-β1 codon 263 is located close to the region where the TGF-β1 dimerization interacts, indicating the TGF-β1 codon 263 variant may affect the secretion of TGF-β1 by altering its dimerization. Together, our findings provide new insights into the immune regulator gene TGF-β1 polymorphisms as susceptibility factors for CLD occurrence and regulators for TGF-β1 expression, which have implications for the regulation of immune factors during CLD development.
Collapse
|
11
|
Lee JH, Massagué J. TGF-β in Developmental and Fibrogenic EMTs. Semin Cancer Biol 2022; 86:136-145. [PMID: 36183999 PMCID: PMC10155902 DOI: 10.1016/j.semcancer.2022.09.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022]
Abstract
TGF-β plays a prominent role as an inducer of epithelial-mesenchymal transitions (EMTs) during development and wound healing and in disease conditions such as fibrosis and cancer. During these processes EMT occurs together with changes in cell proliferation, differentiation, communication, and extracellular matrix remodeling that are orchestrated by multiple signaling inputs besides TGF-β. Chief among these inputs is RAS-MAPK signaling, which is frequently required for EMT induction by TGF-β. Recent work elucidated the molecular basis for the cooperation between the TGF-β-SMAD and RAS-MAPK pathways in the induction of EMT in embryonic, adult and carcinoma epithelial cells. These studies also provided direct mechanistic links between EMT and progenitor cell differentiation during gastrulation or intra-tumoral fibrosis during cancer metastasis. These insights illuminate the nature of TGF-β driven EMTs as part of broader processes during development, fibrogenesis and metastasis.
Collapse
Affiliation(s)
- Jun Ho Lee
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
12
|
Wick KD, Fang X, Maishan M, Matsumoto S, Spottiswoode N, Sarma A, Simoneau C, Khakoo M, Langelier C, Calfee CS, Gotts JE, Matthay MA. Impact of e-cigarette aerosol on primary human alveolar epithelial type 2 cells. Am J Physiol Lung Cell Mol Physiol 2022; 323:L152-L164. [PMID: 35670478 PMCID: PMC9559034 DOI: 10.1152/ajplung.00503.2021] [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: 12/14/2021] [Revised: 05/03/2022] [Accepted: 06/01/2022] [Indexed: 11/22/2022] Open
Abstract
Electronic cigarettes (e-cigarettes) are designed to simulate combustible cigarette smoking and to aid in smoking cessation. Although the number of e-cigarette users has been increasing, the potential health impacts and biological effects of e-cigarettes are still not fully understood. Previous research has focused on the biological effects of e-cigarettes on lung cancer cell lines and distal airway epithelial cells; however, there have been few published studies on the effect of e-cigarettes on primary lung alveolar epithelial cells. The primary purpose of this study was to investigate the direct effect of e-cigarette aerosol on primary human lung alveolar epithelial type 2 (AT2) cells, both alone and in the presence of viral infection. The Melo-3 atomizer caused direct AT2 cell toxicity, whereas the more popular Juul pod's aerosol did not have a detectable cytotoxic effect on AT2 cells. Juul nicotine aerosol also did not increase short-term susceptibility to viral infection. However, 3 days of exposure upregulated genes central to the generation of reactive oxygen species, lipid peroxidation, and carcinogen metabolism and downregulated key innate immune system genes related to cytokine and chemokine signaling. These findings have implications for the potentially injurious impact of long-term use of popular low-power e-cigarette pods on the human alveolar epithelium. Gene expression data might be an important endpoint for evaluating the potential harmful effects of vaping devices that do not cause overt toxicity.
Collapse
Affiliation(s)
- Katherine D Wick
- Cardiovascular Research Institute, University of California, San Francisco, California
| | - Xiaohui Fang
- Cardiovascular Research Institute, University of California, San Francisco, California
| | - Mazharul Maishan
- Cardiovascular Research Institute, University of California, San Francisco, California
| | - Shotaro Matsumoto
- Cardiovascular Research Institute, University of California, San Francisco, California
| | - Natasha Spottiswoode
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, California
| | - Aartik Sarma
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, California
| | - Camille Simoneau
- Gladstone Institutes, University of California, San Francisco, California
| | - Manisha Khakoo
- Cardiovascular Research Institute, University of California, San Francisco, California
| | - Chaz Langelier
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, California
- Chan Zuckerberg Biohub, San Francisco, California
| | - Carolyn S Calfee
- Cardiovascular Research Institute, University of California, San Francisco, California
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, California
| | - Jeffrey E Gotts
- Cardiovascular Research Institute, University of California, San Francisco, California
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California, San Francisco, California
- Department of Medicine, University of California, San Francisco, California
- Department of Anesthesia, University of California, San Francisco, California
| |
Collapse
|
13
|
Kara B, Uyguner O, Maraş Genç H, İşlek EE, Kasap M, Toksoy G, Akpınar G, Uyur Yalçın E, Anık Y, Üstek D. BEND4 as a Candidate Gene for an Infection-Induced Acute Encephalopathy Characterized by a Cyst and Calcification of the Pons and Cerebellar Atrophy. Mol Syndromol 2021; 13:12-22. [DOI: 10.1159/000517541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/01/2021] [Indexed: 11/19/2022] Open
Abstract
Three siblings born to Turkish parents from the same village had normal brain development until acute neurological deterioration between 12 months and 8 years of age. Consequent loss of all acquired motor, social, and language functions following infections was associated with a pontine cyst, calcification, and cerebellar atrophy. Exome sequencing revealed a homozygous c.1297G>A (p.Gly433Ser) alteration in <i>BEND4</i>, which was predicted to be deleterious in in silico analysis tools and segregated in multiple affected individuals in the family. <i>BEND4</i> has not been associated with any existing disease. Immunofluorescence microscopy analysis of wild-type and mutant BEND4 expressing Vero cells showed nuclear and cytoplasmic localization. Wild-type BEND4 displayed a network-like distribution, whereas mutant BEND4 showed a juxtanuclear distribution pattern. Differential proteome analysis of Vero cells expressing BEND4 revealed that mutant BEND4 expression caused selective increase in reticulocalbin-1 and endoplasmic reticulum resident protein-29. Both proteins are associated with the endoplasmic reticulum and are primarily involved in protein processing and folding pathways. Any defect or stress in protein folding creates stress on cells and may cause chronic damage. This is the first study showing that pathogenic <i>BEND4</i> variants may lead to an infection-induced acute necrotizing encephalopathy as demonstrated in characteristic neuroimaging findings.
Collapse
|
14
|
Lei M, Zhao K, Hua W, Wang K, Li S, Wu X, Yang C. An in vivo study of the effect of c-Jun on intervertebral disc degeneration in rats. Bioengineered 2021; 12:4320-4330. [PMID: 34308759 PMCID: PMC8806816 DOI: 10.1080/21655979.2021.1946459] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Intervertebral disc degeneration (IDD) has been well-recognized as one of the causes of vast lower back pain. The objective of the current study intends to elucidate the influence and regulatory molecular mechanisms of c-Jun on IDD. This study established an IDD model of Sprague-Dawley (SD) rats by needle puncture. An LV5-c-Jun lentiviral vector was constructed and injected into rats’ intervertebral disc (IVD) tissue to increase the c-Jun expression following the establishment of modeling. The pathological changes of IVD tissue structure and collagen fibers were visualized following the processes of hematoxylin-eosin (HE) staining method and transmission electron microscopy. Real-time PCR, western blot, immunohistochemistry, and ELISA assays were performed to detect the expression levels of TGF-β, TIMP-3, COL2A1, and inflammatory cytokines. The collagen fibers were arranged in parallel and the surface was smooth after c-Jun overexpression, whereas the collagen fibers in the control group were disorderly arranged with a rough surface. The findings indicated that c-Jun was responsible for upregulating expression levels of TGF-β, TIMP-3, and COL2A1 in the mRNA and proteins, but simultaneously downregulating expression levels of inflammatory factors IL-1β, IL-17, IL-6, and TNF-α. c-Jun overexpression produced a positive effect on IDD, inhibited inflammatory response in vivo, and might delay the degeneration of IVD. Thus, c-Jun may act as a novel potential agent in treating IDD.
Collapse
Affiliation(s)
- Ming Lei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kangcheng Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenbin Hua
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinghuo Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Motizuki M, Koinuma D, Yokoyama T, Itoh Y, Omata C, Miyazono K, Saitoh M, Miyazawa K. TGF-β-induced cell motility requires downregulation of ARHGAPs to sustain Rac1 activity. J Biol Chem 2021; 296:100545. [PMID: 33741342 PMCID: PMC8079281 DOI: 10.1016/j.jbc.2021.100545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/04/2021] [Accepted: 03/15/2021] [Indexed: 01/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) signaling promotes cancer progression. In particular, the epithelial-mesenchymal transition (EMT) induced by TGF-β is considered crucial to the malignant phenotype of cancer cells. Here, we report that the EMT-associated cellular responses induced by TGF-β are mediated by distinct signaling pathways that diverge at Smad3. By expressing chimeric Smad1/Smad3 proteins in SMAD3 knockout A549 cells, we found that the β4 region in the Smad3 MH1 domain is essential for TGF-β-induced cell motility, but is not essential for other EMT-associated responses including epithelial marker downregulation. TGF-β was previously reported to enhance cell motility by activating Rac1 via phosphoinositide 3-kinase. Intriguingly, TGF-β-dependent signaling mediated by Smad3's β4 region causes the downregulation of multiple mRNAs that encode GTPase activating proteins that target Rac1 (ARHGAPs), thereby attenuating Rac1 inactivation. Therefore, two independent pathways downstream of TGF-β type I receptor contribute cooperatively to sustained Rac1 activation, thereby leading to enhanced cell motility.
Collapse
Affiliation(s)
- Mitsuyoshi Motizuki
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Daizo Koinuma
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takashi Yokoyama
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yuka Itoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Chiho Omata
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masao Saitoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan; Center for Medical Education and Science, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Keiji Miyazawa
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.
| |
Collapse
|
16
|
AP-1 and TGFß cooperativity drives non-canonical Hedgehog signaling in resistant basal cell carcinoma. Nat Commun 2020; 11:5079. [PMID: 33033234 PMCID: PMC7546632 DOI: 10.1038/s41467-020-18762-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/01/2020] [Indexed: 12/17/2022] Open
Abstract
Tumor heterogeneity and lack of knowledge about resistant cell states remain a barrier to targeted cancer therapies. Basal cell carcinomas (BCCs) depend on Hedgehog (Hh)/Gli signaling, but can develop mechanisms of Smoothened (SMO) inhibitor resistance. We previously identified a nuclear myocardin-related transcription factor (nMRTF) resistance pathway that amplifies noncanonical Gli1 activity, but characteristics and drivers of the nMRTF cell state remain unknown. Here, we use single cell RNA-sequencing of patient tumors to identify three prognostic surface markers (LYPD3, TACSTD2, and LY6D) which correlate with nMRTF and resistance to SMO inhibitors. The nMRTF cell state resembles transit-amplifying cells of the hair follicle matrix, with AP-1 and TGFß cooperativity driving nMRTF activation. JNK/AP-1 signaling commissions chromatin accessibility and Smad3 DNA binding leading to a transcriptional program of RhoGEFs that facilitate nMRTF activity. Importantly, small molecule AP-1 inhibitors selectively target LYPD3+/TACSTD2+/LY6D+ nMRTF human BCCs ex vivo, opening an avenue for improving combinatorial therapies.
Collapse
|
17
|
SMAD-oncoprotein interplay: Potential determining factors in targeted therapies. Biochem Pharmacol 2020; 180:114155. [DOI: 10.1016/j.bcp.2020.114155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022]
|
18
|
Lei M, Wang K, Li S, Zhao K, Hua W, Wu X, Yang C. The c-Jun signaling pathway has a protective effect on nucleus pulposus cells in patients with intervertebral disc degeneration. Exp Ther Med 2020; 20:123. [PMID: 33005249 PMCID: PMC7523272 DOI: 10.3892/etm.2020.9251] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 07/01/2020] [Indexed: 12/17/2022] Open
Abstract
Among a range of diverse clinical symptoms, intervertebral disc degeneration (IDD) contributes mostly to the onset of lower back pain. The present study aimed to investigate the effects of c-Jun on nucleus pulposus (NP) cells of IDD and its regulation on molecular mechanisms. Intervertebral disc (IVD) tissues were collected from patients suffering from IDD disease, and NP cells were subsequently isolated and cultured. By overexpressing c-Jun in NP cells, expression levels of mRNAs and proteins of IDD-related genes and inflammatory cytokines were subjected to reverse transcription-quantitative PCR, western blot and ELISA assays. Additional transforming growth factor-β (TGF-β) antibodies were administrated to suppress the function of TGF-β. Cell proliferation and apoptosis were determined via Cell Counting Kit-8 and TUNEL assays, respectively. The results demonstrated that the overexpression of c-Jun robustly upregulated both mRNA and protein expression of TGF-β, TIMP metallopeptidase inhibitor 3, aggrecan and collagen type II alpha 1 chain and simultaneously downregulated the expression of the inflammatory cytokines TNF-α, interleukin (IL)-1β, IL-6 and IL-17. Furthermore, following c-Jun overexpression, survival rates of NP cells were increased while apoptosis rates were decreased. However, the addition of a TGF-β antibody significantly promoted apoptosis and restricted cell survival, which differed from the results of the c-Jun overexpression group. The present study hypothesized therefore that c-Jun may positively regulate TGF-β expression within NP cells of IDD, which could promote the proliferation of IDD-NP cells and accelerate cell viability via reducing apoptosis and the inflammatory response.
Collapse
Affiliation(s)
- Ming Lei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Kun Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shuai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Kangcheng Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wenbin Hua
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xinghuo Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
19
|
Aragón E, Wang Q, Zou Y, Morgani SM, Ruiz L, Kaczmarska Z, Su J, Torner C, Tian L, Hu J, Shu W, Agrawal S, Gomes T, Márquez JA, Hadjantonakis AK, Macias MJ, Massagué J. Structural basis for distinct roles of SMAD2 and SMAD3 in FOXH1 pioneer-directed TGF-β signaling. Genes Dev 2019; 33:1506-1524. [PMID: 31582430 PMCID: PMC6824466 DOI: 10.1101/gad.330837.119] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/03/2019] [Indexed: 01/08/2023]
Abstract
TGF-β receptors phosphorylate SMAD2 and SMAD3 transcription factors, which then form heterotrimeric complexes with SMAD4 and cooperate with context-specific transcription factors to activate target genes. Here we provide biochemical and structural evidence showing that binding of SMAD2 to DNA depends on the conformation of the E3 insert, a structural element unique to SMAD2 and previously thought to render SMAD2 unable to bind DNA. Based on this finding, we further delineate TGF-β signal transduction by defining distinct roles for SMAD2 and SMAD3 with the forkhead pioneer factor FOXH1 as a partner in the regulation of differentiation genes in mouse mesendoderm precursors. FOXH1 is prebound to target sites in these loci and recruits SMAD3 independently of TGF-β signals, whereas SMAD2 remains predominantly cytoplasmic in the basal state and set to bind SMAD4 and join SMAD3:FOXH1 at target promoters in response to Nodal TGF-β signals. The results support a model in which signal-independent binding of SMAD3 and FOXH1 prime mesendoderm differentiation gene promoters for activation, and signal-driven SMAD2:SMAD4 binds to promoters that are preloaded with SMAD3:FOXH1 to activate transcription.
Collapse
Affiliation(s)
- Eric Aragón
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Qiong Wang
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Yilong Zou
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Sophie M Morgani
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Lidia Ruiz
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | | | - Jie Su
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Carles Torner
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Lin Tian
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Jing Hu
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Weiping Shu
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Saloni Agrawal
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Tiago Gomes
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | | | | | - Maria J Macias
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain.,ICREA, 08010 Barcelona, Spain
| | - Joan Massagué
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
20
|
Sundqvist A, Morikawa M, Ren J, Vasilaki E, Kawasaki N, Kobayashi M, Koinuma D, Aburatani H, Miyazono K, Heldin CH, van Dam H, Ten Dijke P. JUNB governs a feed-forward network of TGFβ signaling that aggravates breast cancer invasion. Nucleic Acids Res 2019; 46:1180-1195. [PMID: 29186616 PMCID: PMC5814809 DOI: 10.1093/nar/gkx1190] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 11/19/2017] [Indexed: 12/15/2022] Open
Abstract
It is well established that transforming growth factor-β (TGFβ) switches its function from being a tumor suppressor to a tumor promoter during the course of tumorigenesis, which involves both cell-intrinsic and environment-mediated mechanisms. We are interested in breast cancer cells, in which SMAD mutations are rare and interactions between SMAD and other transcription factors define pro-oncogenic events. Here, we have performed chromatin immunoprecipitation (ChIP)-sequencing analyses which indicate that the genome-wide landscape of SMAD2/3 binding is altered after prolonged TGFβ stimulation. De novo motif analyses of the SMAD2/3 binding regions predict enrichment of binding motifs for activator protein (AP)1 in addition to SMAD motifs. TGFβ-induced expression of the AP1 component JUNB was required for expression of many late invasion-mediating genes, creating a feed-forward regulatory network. Moreover, we found that several components in the WNT pathway were enriched among the late TGFβ-target genes, including the invasion-inducing WNT7 proteins. Consistently, overexpression of WNT7A or WNT7B enhanced and potentiated TGFβ-induced breast cancer cell invasion, while inhibition of the WNT pathway reduced this process. Our study thereby helps to explain how accumulation of pro-oncogenic stimuli switches and stabilizes TGFβ-induced cellular phenotypes of epithelial cells.
Collapse
Affiliation(s)
- Anders Sundqvist
- Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Masato Morikawa
- Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Jiang Ren
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Eleftheria Vasilaki
- Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Natsumi Kawasaki
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Mai Kobayashi
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Daizo Koinuma
- Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan
| | - Kohei Miyazono
- Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-751 23 Uppsala, Sweden.,Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Carl-Henrik Heldin
- Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Hans van Dam
- Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Peter Ten Dijke
- Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-751 23 Uppsala, Sweden.,Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
21
|
Genomic Observations of a Rare/Pathogenic SMAD3 Variant in Loeys⁻Dietz Syndrome 3 Confirmed by Protein Informatics and Structural Investigations. ACTA ACUST UNITED AC 2019; 55:medicina55050137. [PMID: 31096651 PMCID: PMC6571799 DOI: 10.3390/medicina55050137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/29/2019] [Accepted: 05/10/2019] [Indexed: 12/12/2022]
Abstract
Background and objectives: Loeys–Dietz syndrome 3, also known as aneurysms-–osteoarthritis syndrome, is an autosomal dominant genetic connective tissue disease caused by pathogenic variants in SMAD3, a transcription factor involved in TGF-β signaling. This disorder is characterized by early-onset osteoarthritis and arterial aneurysms. Common features include scoliosis, uvula abnormalities, striae, and velvety skin. Materials and Methods: The pathogenicity of a variant of uncertain significance in the SMAD3 gene was evaluated (variant c.220C > T) through personalized protein informatics and molecular studies. Results: The case of a 44-year-old male, who was originally presumed to have Marfan syndrome, is presented. An expanded gene panel determined the probable cause to be a variant in SMAD3, c.220C > T (p.R74W). His case was complicated by a history of stroke, but his phenotype was otherwise characteristic for Loeys–Dietz syndrome 3. Conclusion: This case emphasizes the importance of comprehensive genetic testing to evaluate patients for connective tissue disorders, as well as the potential benefit of utilizing a protein informatics platform for the assessment of variant pathogenicity.
Collapse
|
22
|
David CJ, Massagué J. Contextual determinants of TGFβ action in development, immunity and cancer. Nat Rev Mol Cell Biol 2018; 19:419-435. [PMID: 29643418 DOI: 10.1038/s41580-018-0007-0] [Citation(s) in RCA: 575] [Impact Index Per Article: 82.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Few cell signals match the impact of the transforming growth factor-β (TGFβ) family in metazoan biology. TGFβ cytokines regulate cell fate decisions during development, tissue homeostasis and regeneration, and are major players in tumorigenesis, fibrotic disorders, immune malfunctions and various congenital diseases. The effects of the TGFβ family are mediated by a combinatorial set of ligands and receptors and by a common set of receptor-activated mothers against decapentaplegic homologue (SMAD) transcription factors, yet the effects can differ dramatically depending on the cell type and the conditions. Recent progress has illuminated a model of TGFβ action in which SMADs bind genome-wide in partnership with lineage-determining transcription factors and additionally integrate inputs from other pathways and the chromatin to trigger specific cellular responses. These new insights clarify the operating logic of the TGFβ pathway in physiology and disease.
Collapse
Affiliation(s)
- Charles J David
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Tsinghua University School of Medicine, Department of Basic Sciences, Beijing, China
| | - Joan Massagué
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
23
|
Integrative transcriptomics and proteomics analysis of longissimus dorsi muscles of Canadian double-muscled Large White pigs. Gene 2016; 577:14-23. [DOI: 10.1016/j.gene.2015.11.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/28/2015] [Accepted: 11/09/2015] [Indexed: 11/23/2022]
|
24
|
Kuo KK, Jian SF, Li YJ, Wan SW, Weng CC, Fang K, Wu DC, Cheng KH. Epigenetic inactivation of transforming growth factor-β1 target gene HEYL, a novel tumor suppressor, is involved in the P53-induced apoptotic pathway in hepatocellular carcinoma. Hepatol Res 2015; 45:782-93. [PMID: 25179429 DOI: 10.1111/hepr.12414] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 02/08/2023]
Abstract
AIM Hairy/enhancer-of-split related with YRPW motif-like (HEYL) protein was first identified as a transcriptional repressor. It is a downstream gene of the Notch and transforming growth factor-β pathways. Little is known about its role in the pathogenesis of hepatocellular carcinoma (HCC). METHODS Eighty surgically resected paired HCC and adjacent non-cancerous tissues were analyzed for HEYL expression by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry (IHC). HCC cells were transfected with pHEYL-EGFP vector to overexpress the HEYL gene or infected with specific shHEYL lentiviral vector to silence HEYL gene expression. HEYL expressional analysis and functional characterization were assessed by 3-(4 5-dimethylthiazol-2-yl)-2 5-diphenyltetrazolium bromide assays, flow cytometry, RT-qPCR, western blotting and methylation-specific PCR. RESULTS We determined that HEYL expression was inactivated in more than 75% of HCC. In addition, overexpression of HEYL in SK-Hep 1 cells caused apoptosis by the cleavage of caspase 3 and poly (ADP-ribose) polymerase. We discovered that HEYL apoptosis was preceded by serine 15 phosphorylation and accumulation of P53. Molecular analysis revealed that HEYL overexpression led to increased p16, p19, p21, p27 and Bad protein expression, and reduced c-Myc, Bcl-2 and Cyclin B1 expression. Epigenetic silencing of HEYL expression by DNA hypermethylation in HCC directly correlated with loss of HEYL expression in HCC. CONCLUSION HEYL is frequently downregulated by promoter methylation in HCC. HEYL may be a tumor suppressor of liver carcinogenesis through upregulation of P53 gene expression and activation of P53-mediated apoptosis.
Collapse
Affiliation(s)
- Kung-Kai Kuo
- Department of Surgery, Division of Hepatobiliary Pancreatic Surgery, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Fang Jian
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yi-Jin Li
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Shi-Wei Wan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Ching-Chieh Weng
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - KuanTe Fang
- Department of Research and Development, Eternal Chemical, Kaohsiung, Taiwan
| | - Deng-Chyang Wu
- Division of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Division of Gastroenterology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Kuang-Hung Cheng
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| |
Collapse
|
25
|
Roybal LL, Hambarchyan A, Meadows JD, Barakat NH, Pepa PA, Breen KM, Mellon PL, Coss D. Roles of binding elements, FOXL2 domains, and interactions with cJUN and SMADs in regulation of FSHβ. Mol Endocrinol 2014; 28:1640-55. [PMID: 25105693 DOI: 10.1210/me.2014-1008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We previously identified FOXL2 as a critical component in FSHβ gene transcription. Here, we show that mice deficient in FOXL2 have lower levels of gonadotropin gene expression and fewer LH- and FSH-containing cells, but the same level of other pituitary hormones compared to wild-type littermates, highlighting a role of FOXL2 in the pituitary gonadotrope. Further, we investigate the function of FOXL2 in the gonadotrope cell and determine which domains of the FOXL2 protein are necessary for induction of FSHβ transcription. There is a stronger induction of FSHβ reporter transcription by truncated FOXL2 proteins, but no induction with the mutant lacking the forkhead domain. Specifically, FOXL2 plays a role in activin induction of FSHβ, functioning in concert with activin-induced SMAD proteins. Activin acts through multiple promoter elements to induce FSHβ expression, some of which bind FOXL2. Each of these FOXL2-binding sites is either juxtaposed or overlapping with a SMAD-binding element. We determined that FOXL2 and SMAD4 proteins form a higher order complex on the most proximal FOXL2 site. Surprisingly, two other sites important for activin induction bind neither SMADs nor FOXL2, suggesting additional factors at work. Furthermore, we show that FOXL2 plays a role in synergistic induction of FSHβ by GnRH and activin through interactions with the cJUN component of the AP1 complex that is necessary for GnRH responsiveness. Collectively, our results demonstrate the necessity of FOXL2 for proper FSH production in mice and implicate FOXL2 in integration of transcription factors at the level of the FSHβ promoter.
Collapse
Affiliation(s)
- Lacey L Roybal
- Department of Reproductive Medicine (L.L.R., A.H., J.D.M., P.A.P., K.M.B., P.L.M., D.C.), Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093-0674; Division of Biomedical Sciences (N.H.B., D.C.), School of Medicine, University of California, Riverside; Riverside, California 92521
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Reeves SR, Kolstad T, Lien TY, Elliott M, Ziegler SF, Wight TN, Debley JS. Asthmatic airway epithelial cells differentially regulate fibroblast expression of extracellular matrix components. J Allergy Clin Immunol 2014; 134:663-670.e1. [PMID: 24875618 DOI: 10.1016/j.jaci.2014.04.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 04/06/2014] [Accepted: 04/11/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Airway remodeling might explain lung function decline among asthmatic children. Extracellular matrix (ECM) deposition by human lung fibroblasts (HLFs) is implicated in airway remodeling. Airway epithelial cell (AEC) signaling might regulate HLF ECM expression. OBJECTIVES We sought to determine whether AECs from asthmatic children differentially regulate HLF expression of ECM constituents. METHODS Primary AECs were obtained from well-characterized atopic asthmatic (n = 10) and healthy (n = 10) children intubated during anesthesia for an elective surgical procedure. AECs were differentiated at an air-liquid interface for 3 weeks and then cocultured with HLFs from a healthy child for 96 hours. Collagen I (COL1A1), collagen III (COL3A1), hyaluronan synthase (HAS) 2, and fibronectin expression by HLFs and prostaglandin E2 synthase (PGE2S) expression by AECs were assessed by using RT-PCR. TGF-β1 and TGF-β2 concentrations in media were measured by using ELISA. RESULTS COL1A1 and COL3A1 expression by HLFs cocultured with AECs from asthmatic patients was greater than that by HLFs cocultured with AECs from healthy subjects (2.2-fold, P < .02; 10.8-fold, P < .02). HAS2 expression by HLFs cocultured with AECs from asthmatic patients was 2.5-fold higher than that by HLFs cocultured with AECs from healthy subjects (P < .002). Fibronectin expression by HLFs cocultured with AECs from asthmatic patients was significantly greater than that by HLFs alone. TGF-β2 activity was increased in cocultures of HLFs with AECs from asthmatic patients (P < .05), whereas PGES2 was downregulated in AEC-HLF cocultures (2.2-fold, P < .006). CONCLUSIONS HLFs cocultured with AECs from asthmatic patients showed differential expression of the ECM constituents COL1A1 and COL3A1 and HAS2 compared with HLFs cocultured with AECs from healthy subjects. These findings support a role for altered ECM production in asthmatic airway remodeling, possibly regulated by unbalanced AEC signaling.
Collapse
Affiliation(s)
- Stephen R Reeves
- Division of Pulmonary Medicine, Seattle Children's Hospital, University of Washington, Seattle, Wash; Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | - Tessa Kolstad
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | - Tin-Yu Lien
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | - Molly Elliott
- Division of Pulmonary Medicine, Seattle Children's Hospital, University of Washington, Seattle, Wash; Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | | | | | - Jason S Debley
- Division of Pulmonary Medicine, Seattle Children's Hospital, University of Washington, Seattle, Wash; Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash.
| |
Collapse
|
27
|
Cruz ACC, Silva ML, Caon T, Simões CMO. Addition of bone morphogenetic protein type 2 to ascorbate and β-glycerophosphate supplementation did not enhance osteogenic differentiation of human adipose-derived stem cells. J Appl Oral Sci 2013; 20:628-35. [PMID: 23329244 PMCID: PMC3881851 DOI: 10.1590/s1678-77572012000600007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 09/14/2012] [Indexed: 12/25/2022] Open
Abstract
Bone morphogenetic protein type 2 (BMP-2) is a potent local factor, which promotes
bone formation and has been used as an osteogenic supplement for mesenchymal stem
cells.
Collapse
|
28
|
Lee SY, Yoon J, Lee HS, Hwang YS, Cha SW, Jeong CH, Kim JI, Park JB, Lee JY, Kim S, Park MJ, Dong Z, Kim J. The function of heterodimeric AP-1 comprised of c-Jun and c-Fos in activin mediated Spemann organizer gene expression. PLoS One 2011; 6:e21796. [PMID: 21829441 PMCID: PMC3146467 DOI: 10.1371/journal.pone.0021796] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 06/13/2011] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Activator protein-1 (AP-1) is a mediator of BMP or FGF signaling during Xenopus embryogenesis. However, specific role of AP-1 in activin signaling has not been elucidated during vertebrate development. METHODOLOGY/PRINCIPAL FINDINGS We provide new evidence showing that overexpression of heterodimeric AP-1 comprised of c-jun and c-fos (AP-1(c-Jun/c-Fos)) induces the expression of BMP-antagonizing organizer genes (noggin, chordin and goosecoid) that were normally expressed by high dose of activin. AP-1(c-Jun/c-Fos) enhanced the promoter activities of organizer genes but reduced that of PV.1, a BMP4-response gene. A loss of function study clearly demonstrated that AP-1(c-Jun/c-Fos) is required for the activin-induced organizer and neural gene expression. Moreover, physical interaction of AP-1(c-Jun/c-Fos) and Smad3 cooperatively enhanced the transcriptional activity of goosecoid via direct binding on this promoter. Interestingly, Smad3 mutants at c-Jun binding site failed in regulation of organizer genes, indicating that these physical interactions are specifically necessary for the expression of organizer genes. CONCLUSIONS/SIGNIFICANCE AP-1(c-Jun/c-Fos) plays a specific role in organizer gene expression in downstream of activin signal during early Xenopus embryogenesis.
Collapse
Affiliation(s)
- Sung-Young Lee
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Jaeho Yoon
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| | - Hyun-Shik Lee
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Yoo-Seok Hwang
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sang-Wook Cha
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chul-Ho Jeong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Jong-Il Kim
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| | - Jae-Yong Lee
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| | - SungChan Kim
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| | - Mae Ja Park
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Jaebong Kim
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| |
Collapse
|
29
|
Affiliation(s)
- Harold A. Chapman
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, California 94143;
| |
Collapse
|
30
|
Thackray VG, Mellon PL, Coss D. Hormones in synergy: regulation of the pituitary gonadotropin genes. Mol Cell Endocrinol 2010; 314:192-203. [PMID: 19747958 PMCID: PMC2815122 DOI: 10.1016/j.mce.2009.09.003] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 09/02/2009] [Accepted: 09/02/2009] [Indexed: 11/23/2022]
Abstract
The precise interplay of hormonal influences that governs gonadotropin hormone production by the pituitary includes endocrine, paracrine and autocrine actions of hypothalamic gonadotropin-releasing hormone (GnRH), activin and steroids. However, most studies of hormonal regulation of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) in the pituitary gonadotrope have been limited to analyses of the isolated actions of individual hormones. LHbeta and FSHbeta subunits have distinct patterns of expression during the menstrual/estrous cycle as a result of the integration of activin, GnRH, and steroid hormone action. In this review, we focus on studies that delineate the interplay among these hormones in the regulation of LHbeta and FSHbeta gene expression in gonadotrope cells and discuss how signaling cross-talk contributes to differential expression. We also discuss how recent technological advances will help identify additional factors involved in the differential hormonal regulation of LH and FSH.
Collapse
Affiliation(s)
| | | | - Djurdjica Coss
- To whom the correspondence should be addressed: Djurdjica Coss, Department of Reproductive Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674, Phone: (858) 534-1762, Fax: (858) 534-1438,
| |
Collapse
|
31
|
Liebhart J, Dobek R. Transforming growth factor-beta in the pathogenesis of chronic obstructive pulmonary disease. ACTA ACUST UNITED AC 2009. [DOI: 10.1080/17471060701721985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
32
|
Tsuchiya M, Sharma R, Tye CE, Sugiyama T, Bartlett JD. Transforming growth factor-beta1 expression is up-regulated in maturation-stage enamel organ and may induce ameloblast apoptosis. Eur J Oral Sci 2009; 117:105-12. [PMID: 19320718 DOI: 10.1111/j.1600-0722.2009.00612.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Transforming growth factor-beta1 (TGF-beta1) regulates a variety of cellular responses that are dependent on the developmental stage and on the origins of the cell or the tissue. In mature tissues, and especially in tissues of epithelial origin, TGF-beta1 is generally considered to be a growth inhibitor that may also promote apoptosis. The ameloblast cells of the enamel organ epithelium are adjacent to and responsible for the developing enamel layer on unerupted teeth. Once the enamel layer reaches its full thickness, the tall columnar secretory-stage ameloblasts shorten, and a portion of these maturation-stage ameloblasts become apoptotic. Here we investigate whether TGF-beta1 plays a role in apoptosis of the maturation-stage ameloblasts. We demonstrate in vitro that ameloblast lineage cells are highly susceptible to TGF-beta1-mediated growth arrest and are prone to TGF-beta1-mediated cell death/apoptosis. We also demonstrate in vivo that TGF-beta1 is expressed in the maturation-stage enamel organ at significantly higher levels than in the earlier secretory-stage enamel organ. This increased expression of TGF-beta1 correlates with an increase in expression of the enamel organ immediate-early stress-response gene and with a decrease in the anti-apoptotic Bcl2 : Bax expression ratio. We conclude that TGF-beta1 may play an important role in ameloblast apoptosis during the maturation stage of enamel development.
Collapse
Affiliation(s)
- Masahiro Tsuchiya
- Department of Cytokine Biology, Forsyth Institute, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
33
|
Huang J, Dattilo LK, Rajagopal R, Liu Y, Kaartinen V, Mishina Y, Deng CX, Umans L, Zwijsen A, Roberts AB, Beebe DC. FGF-regulated BMP signaling is required for eyelid closure and to specify conjunctival epithelial cell fate. Development 2009; 136:1741-50. [PMID: 19369394 DOI: 10.1242/dev.034082] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
There are conflicting reports about whether BMP signaling is required for eyelid closure during fetal development. This question was addressed using mice deficient in BMP or TGFbeta signaling in prospective eyelid and conjunctival epithelial cells. Genes encoding two type I BMP receptors, the type II TGFbeta receptor, two BMP- or two TGFbeta-activated R-Smads or the co-Smad Smad4 were deleted from the ocular surface ectoderm using Cre recombinase. Only mice with deletion of components of the BMP pathway had an 'eyelid open at birth' phenotype. Mice lacking Fgf10 or Fgfr2 also have open eyelids at birth. To better understand the pathways that regulate BMP expression and function during eyelid development, we localized BMPs and BMP signaling intermediates in Fgfr2 and Smad4 conditional knockout (CKO) mice. We found that Fgfr2 was required for the expression of Bmp4, the normal distribution of Shh signaling and for preserving the differentiation of the conjunctival epithelium. FGF signaling also promoted the expression of the Wnt antagonist Sfrp1 and suppressed Wnt signaling in the prospective eyelid epithelial cells, independently of BMP function. Transcripts encoding Foxc1 and Foxc2, which were previously shown to be necessary for eyelid closure, were not detectable in Smad4(CKO) animals. c-Jun, another key regulator of eyelid closure, was present and phosphorylated in eyelid periderm cells at the time of fusion, but failed to translocate to the nucleus in the absence of BMP function. Smad4(CKO) mice also showed premature differentiation of the conjunctival epithelium, conjunctival hyperplasia and the acquisition of epidermal characteristics, including formation of an ectopic row of hair follicles in place of the Meibomian glands. A second row of eyelashes is a feature of human lymphedema-distichiasis syndrome, which is associated with mutations in FOXC2.
Collapse
Affiliation(s)
- Jie Huang
- Department of Ophthalmology and Visual Sciences, Washington University, St Louis, MO 63130, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Burgess JK. The role of the extracellular matrix and specific growth factors in the regulation of inflammation and remodelling in asthma. Pharmacol Ther 2009; 122:19-29. [PMID: 19141302 DOI: 10.1016/j.pharmthera.2008.12.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 12/17/2008] [Indexed: 12/12/2022]
Abstract
Asthma is a disease characterised by persistent inflammation and structural changes in the airways, referred to as airway remodelling. The mechanisms underlying these processes may be interdependent or they may be separate processes that are driven by common factors. The levels of a variety of growth factors (including transforming growth factor beta, granulocyte macrophage colony stimulating factor, and vascular endothelial growth factor) are known to be changed in the asthmatic airway. These and other growth factors can contribute to the development and persistence of inflammation and remodelling. One of the prominent features of the structural changes of the airways is the increased deposition and alterations in the composition of the extracellular matrix proteins. These proteins include fibronectin, many different collagen types and hyaluronan. There is a dynamic relationship between the extracellular matrix proteins and the airway mesenchymal cells such that the changes in the extracellular matrix proteins can also contribute to the persistence of inflammation and the airway remodelling. This review aims to summarise the role growth factors and extracellular matrix proteins play in the regulation of inflammation and airway remodelling in the asthmatic airway.
Collapse
Affiliation(s)
- Janette K Burgess
- Discipline of Pharmacology, The University of Sydney, Woolcock Institute of Medical Research and the Cooperative Research Centre for Asthma and Airways, Sydney, NSW Australia.
| |
Collapse
|
35
|
TGFbeta1 antagonistic peptides inhibit TGFbeta1-dependent angiogenesis. Biochem Pharmacol 2008; 77:813-25. [PMID: 19041849 DOI: 10.1016/j.bcp.2008.10.036] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 10/24/2008] [Accepted: 10/31/2008] [Indexed: 11/21/2022]
Abstract
The role of transforming growth factor beta (TGFbeta) in tumor promotion and in angiogenesis is context-dependent. While TGFbeta prevents tumor growth and angiogenesis in early phases of tumor development, evidence is accumulating about its pro-angiogenic and tumor promotion activities in late-stages of tumor progression. Here we have studied, in an experimental context previously reported to disclose the pro-angiogenic effects of TGFbeta, the blocking activity of TGFbeta antagonist peptides. In agreement with previous results, we have observed that TGFbeta exerts a powerful pro-angiogenic activity on human normal dermal microvascular endothelial cells (MVEC), by promoting invasion and capillary morphogenesis in Matrigel. No apoptotic activity of TGFbeta was observed. By RT-PCR we have shown that TGFbeta up-regulates expression not only of plasminogen activator inhibitor type-1 (PAI-1), but also of the urokinase-type plasminogen activator receptor (uPAR), whose inhibition by specific antibodies blunted the TGFbeta angiogenic response in vitro. The SMAD2/3 and FAK signaling pathways were activated by TGFbeta in MVEC, as an early and late response, respectively. The use of two different TGFbeta1 antagonist peptides, derived from TGFbeta type III receptor sequence and 15-mer phage display technology, inhibited the signaling and pro-angiogenic response in vitro, as well as uPAR and PAI-1 up-regulation of MVEC following TGFbeta challenge. The anti-angiogenic properties of both inhibitors were evident also in the in vivo TGFbeta Matrigel Sponge Assay. These results may be relevant to develop a potentially fruitful strategy for the therapy of late-stage-associated tumor angiogenesis.
Collapse
|
36
|
Walsh MF, Ampasala DR, Hatfield J, Vander Heide R, Suer S, Rishi AK, Basson MD. Transforming growth factor-beta stimulates intestinal epithelial focal adhesion kinase synthesis via Smad- and p38-dependent mechanisms. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:385-99. [PMID: 18583311 DOI: 10.2353/ajpath.2008.070729] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Focal adhesion kinase (FAK) regulates cell migration, proliferation, and apoptosis. FAK protein is reduced at the edge of migrating gut epithelial sheets in vitro, but it has not been characterized in restitutive gut mucosa in vivo. Here we show that FAK and activated phospho-FAK (FAK(397)) immunoreactivity was lower in epithelial cells immediately adjacent to human gastric and colonic ulcers in vivo, but dramatically increased in epithelia near the ulcers, possibly reflecting stimulation by growth factors absent in vitro. Transforming growth factor (TGF)-beta, but not fibroblast growth factor, platelet-derived growth factor, or vascular endothelial growth factor, increased FAK levels in Caco-2 and IEC-6 cells. Epithelial immunoreactivity to TGF-beta and phospho-Smad3 was also higher near the ulcers, varying in parallel with FAK. The TGF-beta receptor antagonist SB431542 completely blocked TGF-beta-induced Smad2/3 and p38 activation in IEC-6 cells. SB431542, the p38 antagonist SB203580, and siRNA-mediated reduction of Smad2 and p38alpha prevented TGF-beta stimulation of both FAK transcription and translation (as measured via a FAK promoter-luciferase construct). FAK(397) levels were directly related to total FAK protein expression. Although gut epithelial motility is associated with direct inhibition of FAK protein adjacent to mucosal wounds, TGF-beta may increase FAK protein near but not bordering mucosal ulcers via Smad2/3 and p38 signals. Our results show that regulation of FAK expression may be as important as FAK phosphorylation in critically influencing gut epithelial cell migration after mucosal injury.
Collapse
Affiliation(s)
- Mary F Walsh
- Departments of Surgery and Pathology, John D. Dingell VA Medical Center, Wayne State University, 4646 John R Detroit, MI 48201-1932, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Li T, Ma H, Chiang JYL. TGFbeta1, TNFalpha, and insulin signaling crosstalk in regulation of the rat cholesterol 7alpha-hydroxylase gene expression. J Lipid Res 2008; 49:1981-9. [PMID: 18511845 DOI: 10.1194/jlr.m800140-jlr200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The TGFbeta1/Smad pathway plays a critical role in cholestasis and liver fibrosis. Previous studies show that TGFbeta1, TNFalpha, and insulin inhibit cholesterol 7alpha-hydroxylase (CYP7A1) gene transcription and bile acid synthesis in human hepatocytes. In this study, we investigated insulin, TGFbeta1, and TNFalpha regulation of rat Cyp7a1 gene transcription. In contrast to inhibition of human CYP7A1 gene transcription, TGFbeta1 stimulates rat Cyp7a1 reporter activity. Smad3, FoxO1, and HNF4alpha synergistically stimulated rat Cyp7a1 gene transcription. Mutations of the Smad3, FoxO1, or HNF4alpha binding site attenuated the rat Cyp7a1 promoter activity. Furthermore, TNFalpha and cJun attenuated TGFbeta1 stimulation of rat Cyp7a1. Insulin or adenovirus-mediated expression of constitutively active AKT1 inhibited FoxO1 and Smad3 synergy. In streptozotocin-induced diabetic rats, Cyp7a1 mRNA expression levels were induced and insulin attenuated CYP7A1 mRNA levels. Chromatin immunoprecipitation assay showed that FoxO1 binding to Cyp7a1 chromatin was increased in diabetic rat livers and insulin reduced FoxO1 binding. These results suggest a mechanistic basis for induction of Cyp7a1 activity and bile acid synthesis in cholestatic rats and in diabetic rats. The crosstalk of insulin, TGFbeta and TNFalpha signaling pathways may regulate bile acid synthesis and lipid homeostasis in diabetes, fatty liver disease, and liver fibrosis.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Integrative Medical Sciences, Northeastern Ohio Universities Colleges of Medicine and Pharmacy, Rootstown, OH 44272, USA
| | | | | |
Collapse
|
38
|
Arnott JA, Zhang X, Sanjay A, Owen TA, Smock SL, Rehman S, DeLong WG, Safadi FF, Popoff SN. Molecular requirements for induction of CTGF expression by TGF-beta1 in primary osteoblasts. Bone 2008; 42:871-85. [PMID: 18314002 PMCID: PMC2430079 DOI: 10.1016/j.bone.2008.01.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 11/30/2007] [Accepted: 01/03/2008] [Indexed: 01/05/2023]
Abstract
Connective tissue growth factor (CTGF/CCN2) is a cysteine rich, extracellular matrix protein that acts as an anabolic growth factor to regulate osteoblast differentiation and function. In osteoblasts, CTGF is induced by TGF-beta1 where it acts as a downstream mediator of TGF-beta1 induced matrix production. The molecular mechanisms that control CTGF induction by TGF-beta1 in osteoblasts are not known. To assess the role of individual Smads in mediating the induction of CTGF by TGF-beta1, we used specific Smad siRNAs to block Smad expression. These studies demonstrated that Smads 3 and 4, but not Smad 2, are required for TGF-beta1 induced CTGF promoter activity and expression in osteoblasts. Since the activation of MAPKs (Erk, Jnk and p38) by TGF-beta1 is cell type specific, we were interested in determining the role of individual MAPKs in TGF-beta1 induction of CTGF promoter activity and expression. Using dominant negative (DN) mutants for Erk, Jnk and p38, we demonstrated that the expression of DN-Erk caused a significant inhibition of TGF-beta1 induced CTGF promoter activity. In contrast, the expression of DN-p38 or DN-Jnk failed to inhibit activation of CTGF promoter activity. To confirm the vital role of Erk, we used the Erk inhibitor (PD98059) to block its activation, demonstrating that it prevented TGF-beta1 activation of the CTGF promoter and up-regulation of CTGF expression in osteoblasts. Since Src can also act as a downstream signaling effector for TGF-beta in some cell types, we determined its role in TGF-beta1 induction of CTGF in osteoblasts. Treatment of osteoblasts with a Src family kinase inhibitor, PP2, or the expression of two independent kinase-dead Src mutant constructs caused significant inhibition of TGF-beta1 induced CTGF promoter activity and expression. Additionally, blocking Src activation prevented Erk activation by TGF-beta1 demonstrating a role for Src as an upstream mediator of Erk in regulating CTGF expression in osteoblasts. To investigate the involvement of the TGF-beta1 response element (TRE) and the SMAD binding element (SBE) in CTGF induction, we cloned the rat CTGF proximal promoter (-787 to +1) containing the TRE and SBE motifs into a pGL3-Luciferase reporter construct. Using a combination of CTGF promoter deletion constructs and site-directed mutants, we demonstrated the unique requirement of both the TRE and SBE for CTGF induction by TGF-beta1 in osteoblasts. Electro-mobility shift assays using specific probes containing the TRE, SBE or both showed TGF-beta1 inducible complexes that can be ablated by mutation of the respective motif, confirming their requirement for TGF-beta1 induced CTGF promoter activity. In conclusion, these studies demonstrate that CTGF induction by TGF-beta1 in osteoblasts involves Smads 3 and 4, the Erk and Src signaling pathways, and requires both the TRE and SBE motifs in the CTGF proximal promoter.
Collapse
Affiliation(s)
- J A Arnott
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Increased CCN2 Transcription in Keloid Fibroblasts Requires Cooperativity Between AP-1 and SMAD Binding Sites. Ann Surg 2007; 246:886-95. [DOI: 10.1097/sla.0b013e318070d54f] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
40
|
Coss D, Hand CM, Yaphockun KKJ, Ely HA, Mellon PL. p38 mitogen-activated protein kinase is critical for synergistic induction of the FSH(beta) gene by gonadotropin-releasing hormone and activin through augmentation of c-Fos induction and Smad phosphorylation. Mol Endocrinol 2007; 21:3071-86. [PMID: 17823303 PMCID: PMC2932484 DOI: 10.1210/me.2007-0247] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
GnRH and activin independently and synergistically activate transcription of the FSH beta-subunit gene, the subunit that provides specificity and is the limiting factor in the synthesis of the mature hormone. This synergistic interaction, as determined by two-way ANOVA, is specific for FSHbeta and may, therefore, contribute to differential expression of the two gonadotropin hormones, which is critical for the reproductive cycle. We find that the cross-talk between the GnRH and activin signaling pathways occurs at the level of p38 MAPK, because the synergy is dependent on p38 MAPK activity, which is activated by GnRH, and activin cotreatment augments p38 activation by GnRH. Both the Smad and activator protein-1 binding sites on the FSHbeta promoter are necessary and sufficient for synergy. After cotreatment, Smad 3 proteins are more highly phosphorylated on the activin-receptor signaling-dependent residues on the C terminus than with activin treatment alone, and c-Fos is more highly expressed than with GnRH treatment alone. Inhibition of p38 by either of two different inhibitors or a dominant-negative p38 kinase abrogates synergy on FSHbeta expression, reduces c-Fos induction by GnRH, and prevents the further increase in c-Fos levels that occurs with cotreatment. Additionally, p38 is necessary for maximal Smad 3 C-terminal phosphorylation by activin treatment alone and for the further increase caused by cotreatment. Thus, p38 is the pivotal signaling molecule that integrates GnRH and activin interaction on the FSHbeta promoter through higher induction of c-Fos and elevated Smad phosphorylation.
Collapse
Affiliation(s)
- Djurdjica Coss
- Department of Reproductive Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0674, USA
| | | | | | | | | |
Collapse
|
41
|
Farrington DL, Yingling JM, Fill JA, Yan L, Qian YW, Shou J, Wang X, Ehsani ME, Cleverly AL, Daly TM, Lahn M, Konrad RJ, Ray CA. Development and validation of a phosphorylated SMAD ex vivo stimulation assay. Biomarkers 2007; 12:313-30. [PMID: 17453744 DOI: 10.1080/13547500601162441] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Assessing the pharmacodynamics (PD) of a potential therapeutic through the use of a downstream biomarker is essential. This is traditionally performed in the target tissue but limited volume and invasiveness of sampling pose challenges with solid tumours. Currently, there are several small molecule receptor kinase inhibitors and large molecule therapeutic antibodies in clinical trials that interfere with TGFbeta signalling to treat various forms of cancer. With the advent of these new therapies, there is a need for a surrogate tissue that is easily accessible and indicative of tumour response. We propose the use of an ex vivo TGFbeta1 stimulation of peripheral blood mononuclear cells (PBMCs) coupled with the measurement of phosphorylated SMAD2 (Sma/Mothers Against dpp, a downstream transcriptional activator) using a sandwich ELISA. TGFbeta is involved in many different cellular responses, such as proliferation, angiogenesis, migration, invasion and immunomodulation. SMAD2 and SMAD3 are phosphorylated as a result of the canonical cascade through ligand binding and receptor kinase activation. These phosphorylated SMADs (pSMAD) associate with SMAD4, a co-SMAD, and transcriptionally activate TGFbeta-mediated genes. This paper describes the novel method for measuring the downstream effects of inhibiting canonical TGFbeta signalling using ex vivo stimulation of surrogate tissue to predict tumour response. In addition, we present the assay validation rationale and data. This novel, validated assay can be used to gain insight into clinical trials regarding TGFbeta signal modulation by multiple inhibitor platforms for both large and small molecules.
Collapse
MESH Headings
- Activin Receptors, Type I/antagonists & inhibitors
- Animals
- Biomarkers/analysis
- Biomarkers/metabolism
- Blotting, Western
- Cell Line, Tumor
- Data Interpretation, Statistical
- Dose-Response Relationship, Drug
- Drug Evaluation, Preclinical/methods
- Enzyme-Linked Immunosorbent Assay
- Female
- Humans
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Mice
- Mice, Nude
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Neoplasms/pathology
- Phosphorylation/drug effects
- Protein Kinase Inhibitors/pharmacology
- Protein Serine-Threonine Kinases
- Rats
- Rats, Inbred F344
- Receptor, Transforming Growth Factor-beta Type I
- Receptors, Transforming Growth Factor beta/antagonists & inhibitors
- Reproducibility of Results
- Smad Proteins/analysis
- Smad Proteins/metabolism
- Smad2 Protein/analysis
- Smad2 Protein/metabolism
- Smad3 Protein/analysis
- Smad3 Protein/metabolism
- Transforming Growth Factor beta1/pharmacology
- Xenograft Model Antitumor Assays/methods
Collapse
Affiliation(s)
- D L Farrington
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Brown KA, Pietenpol JA, Moses HL. A tale of two proteins: differential roles and regulation of Smad2 and Smad3 in TGF-beta signaling. J Cell Biochem 2007; 101:9-33. [PMID: 17340614 DOI: 10.1002/jcb.21255] [Citation(s) in RCA: 302] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transforming growth factor-beta (TGF-beta) is an important growth inhibitor of epithelial cells, and insensitivity to this cytokine results in uncontrolled cell proliferation and can contribute to tumorigenesis. Smad2 and Smad3 are direct mediators of TGF-beta signaling, however little is known about the selective activation of Smad2 versus Smad3. The Smad2 and Smad3 knockout mouse phenotypes and studies comparing Smad2 and Smad3 activation of TGF-beta target genes, suggest that Smad2 and Smad3 have distinct roles in TGF-beta signaling. The observation that TGF-beta inhibits proliferation of Smad3-null mammary gland epithelial cells, whereas Smad3 deficient fibroblasts are only partially growth inhibited, suggests that Smad3 has a different role in epithelial cells and fibroblasts. Herein, the current understanding of Smad2 and Smad3-mediated TGF-beta signaling and their relative roles are discussed, in addition to potential mechanisms for the selective activation of Smad2 versus Smad3. Since alterations in the TGF-beta signaling pathway play an important role in promoting tumorigenesis and cancer progression, methods for therapeutic targeting of the TGF-beta signaling pathway are being pursued. Determining how Smad2 or Smad3 differentially regulate the TGF-beta response may translate into developing more effective strategies for cancer therapy.
Collapse
Affiliation(s)
- Kimberly A Brown
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | |
Collapse
|
43
|
Makinde T, Murphy RF, Agrawal DK. The regulatory role of TGF-beta in airway remodeling in asthma. Immunol Cell Biol 2007; 85:348-56. [PMID: 17325694 DOI: 10.1038/sj.icb.7100044] [Citation(s) in RCA: 209] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Both structural and inflammatory cells are capable of secreting transforming growth factor (TGF)-beta and expressing TGF-beta receptors. TGF-beta can induce multiple cellular responses including differentiation, apoptosis, survival and proliferation, and has been implicated in the development of several pathogenic conditions including cancer and asthma. Elevated levels of TGF-beta have been reported in the asthmatic airway. TGF-beta binds to its receptor complex and activates multiple pathways involving proteins such as Sma and Mad homologues, phosphatidylinositol-3 kinase and the mitogen-activated protein kinases, leading to the transcription of several genes. Cell type, cellular condition, and microenvironment, all play a role in determining which pathway is activated, which, in turn, is an indication of which gene is to be transcribed. TGF-beta has been shown to induce apoptosis in airway epithelial cells. A possible role for TGF-beta in the regulation of epithelial cell adhesion properties has also been reported. Enhancement of goblet cell proliferation by TGF-beta suggests a role in mucus hyper-secretion. Elevated levels of TGF-beta correlate with subepithelial fibrosis. TGF-beta induces proliferation of fibroblast cells and their differentiation into myofibroblasts and extracellular matrix (ECM) protein synthesis during the development of subepithelial fibrosis. TGF-beta also induces proliferation and survival of and ECM secretion in airway smooth muscle cells (ASMCs), suggesting a possible cause of increased thickness of airway tissues. TGF-beta also induces the production and release of vascular endothelial cell growth factor and plasminogen activator inhibitor, contributing to the vascular remodeling in the asthmatic airway. Blocking TGF-beta activity inhibits epithelial shedding, mucus hyper-secretion, angiogenesis, ASMC hypertrophy and hyperplasia in an asthmatic mouse model. Reduction of TGF-beta production and control of TGF-beta effects would be beneficial in the development of therapeutic intervention for airway remodeling in chronic asthma.
Collapse
Affiliation(s)
- Toluwalope Makinde
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA
| | | | | |
Collapse
|
44
|
Wu Y, Zhang X, Salmon M, Lin X, Zehner ZE. TGFbeta1 regulation of vimentin gene expression during differentiation of the C2C12 skeletal myogenic cell line requires Smads, AP-1 and Sp1 family members. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1773:427-39. [PMID: 17270292 PMCID: PMC1855268 DOI: 10.1016/j.bbamcr.2006.11.017] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Revised: 11/22/2006] [Accepted: 11/27/2006] [Indexed: 10/23/2022]
Abstract
Vimentin exhibits a complex pattern of developmental and tissue-specific expression regulated by such growth factors as TGFbeta1, PDGF, FGF, EGF and cytokines. Vimentin is expressed in the more migratory, mesenchymal cell and its expression is often down-regulated to make way for tissue-specific intermediate filaments proteins such as desmin in muscle. Here, we suggest a mechanism to explain how TGFbeta1 contributes to the up-regulation of vimentin expression while blocking myogenesis. TGFbeta1 binds to serine/threonine kinase receptors resulting in the phosphorylation of Smad2 and Smad3, followed by formation of a heteromeric complex with Smad4. The translocation of this complex to the nucleus modulates transcription of selected genes such as vimentin. However, the vimentin gene lacks a consensus TGFbeta1 response element. By transient transfection analysis of vimentin's various promoter elements fused to the CAT reporter gene, we have determined that tandem AP-1 sites surrounded by GC-boxes are required for TGFbeta1 induction. Mutations within this region eliminated the ability of Smad3 to induce reporter gene expression. DNA precipitation and ChIP assays suggest that c-Jun, c-Fos, Smad3 and Sp1/Sp3 interact over this region, but this interaction changes during myogenesis with TGFbeta1 induction.
Collapse
Affiliation(s)
- Yongzhong Wu
- Department of Biochemistry and the Massey Cancer Center, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond VA 23298-0614
| | - Xueping Zhang
- Department of Biochemistry and the Massey Cancer Center, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond VA 23298-0614
| | - Morgan Salmon
- Department of Biochemistry and the Massey Cancer Center, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond VA 23298-0614
| | - Xia Lin
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030
| | - Zendra E. Zehner
- Department of Biochemistry and the Massey Cancer Center, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond VA 23298-0614
| |
Collapse
|
45
|
Li X, Thyssen G, Beliakoff J, Sun Z. The Novel PIAS-like Protein hZimp10 Enhances Smad Transcriptional Activity. J Biol Chem 2006; 281:23748-56. [PMID: 16777850 DOI: 10.1074/jbc.m508365200] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor beta (TGF-beta) plays critical roles in the control of cell proliferation, differentiation, and apoptosis. Smad proteins are substrates of the TGF-beta type I receptor and are responsible for transducing receptor signals to target genes in the nucleus. The PIAS (protein inhibitor of activated STAT) proteins were originally identified as transcriptional co-regulators of the JAK-STAT pathway. Subsequently, cross-talk between the PIAS proteins and other signaling pathways has been shown to be involved in various cellular processes. Importantly, PIAS proteins modulate TGF-beta signaling by regulating the transcriptional activity of Smad3. In this study we tested whether hZimp10, a novel PIAS-like protein, acts as other PIAS proteins to regulate Smad3-mediated transcription. We show that expression of exogenous hZimp10 enhances the transcriptional activity of Smad3, which appears to be Smad4-dependent and responsive to TGF-beta induction. Furthermore, knockdown of endogenous hZimp10 reduced the transcriptional activity of Smad3. A protein-protein interaction between Smad3 and Smad4 with hZimp10 was identified in glutathione S-transferase-pulldown and co-immunoprecipitation assays. The Miz domain of hZimp10 and the MH2 domains of Smad3 and Smad4 were mapped as the regions responsible for binding. Results from immunostaining assays further demonstrated that Smad3, Smad4, and hZimp10 co-localize within cell nuclei. Finally, we demonstrated that Smad3/4-mediated transcription is significantly impaired in response to TGF-beta induction in Zimp10 null (zimp10-/-) embryonic fibroblasts. Taken together, these results provide the first line of evidence to demonstrate a role for Zimp10 in regulating the TGF-beta/Smad signaling pathway.
Collapse
Affiliation(s)
- Xiaomeng Li
- Department of Urology, Stanford University School of Medicine, Stanford, California 94303-5118, USA
| | | | | | | |
Collapse
|
46
|
Funaba M, Ikeda T, Murakami M, Ogawa K, Nishino Y, Tsuchida K, Sugino H, Abe M. Transcriptional regulation of mouse mast cell protease-7 by TGF-β. ACTA ACUST UNITED AC 2006; 1759:166-70. [PMID: 16730810 DOI: 10.1016/j.bbaexp.2006.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2006] [Revised: 04/07/2006] [Accepted: 04/10/2006] [Indexed: 10/24/2022]
Abstract
Mouse mast cell protease-7 (mmcp-7) is a tryptase predominantly expressed in differentiated connective tissue-type mast cells. Previous study revealed that transforming growth factor-beta (TGF-beta) increases gene transcript of mmcp-7 in mast cells. The present study explored molecular mechanism of the up-regulation of mmcp-7 by TGF-beta. Luciferase-based reporter assays using deletion and point mutations of mmcp-7 promoter showed a critical region spanning nt -126 to -122 relative to the transcriptional start site, a Smad-binding element, for transcriptional activation by the TGF-beta pathway. In addition, a region from nt -104 to -98, a TPA-responsive element, was also necessary for the transactivation. Consistent with the current model for the TGF-beta signaling, Smad4 was required for the transcription of mmcp-7 by Smad3, a signal mediator of TGF-beta. Treatment with TGF-beta in mast cells resulted in the differential gene induction of the AP-1 components, i.e., transient induction of c-fos but not of c-jun and junB. Expression of c-fos further enhanced Smad3 and Smad4-induced transcription of mmcp-7, whereas c-jun expression inhibited the transcription. Our results suggest that TGF-beta stimulates mmcp-7 transcription through the Smad3-Smad4 pathway as well as c-fos induction, and that the AP-1 components distinctly related with the TGF-beta pathway.
Collapse
Affiliation(s)
- Masayuki Funaba
- Laboratory of Nutrition, Azabu University School of Veterinary Medicine, 1-17-71 Fuchinobe, Sagamihara 229-8501, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW The attempt of this review is to bring into focus the potential role of dietary salt intake in progression of chronic kidney disease. RECENT FINDINGS Ongoing work has elucidated a role for dietary salt intake in modulating intrarenal production of transforming growth factor-beta1. The mechanism is independent of angiotensin II and systemic blood pressure and involves activation of vascular endothelium by dietary salt intake with release of this growth factor. In this model, transforming growth factor-beta1 serves an autacoid function by stimulating nitric oxide production by the endothelium. In turn, endothelium-derived nitric oxide modulates production of this growth factor. The model further predicts that individuals who have lost the requisite endothelial cell flexibility to adapt to this environmental stress (a high salt diet) are potentially at increased risk of developing end-organ damage from excess salt intake. Animal and human studies are presented to support this working hypothesis. SUMMARY Overproduction of transforming growth factor-beta1 permits excess biological activity of this important fibrogenic growth factor with subsequent development or acceleration of vascular and kidney damage. In patients with diseases whose pathogenesis is related to excess production of transforming growth factor-beta1, such as chronic allograft nephropathy and diabetic nephropathy, increased salt intake may hasten loss of function, particularly if nitric oxide production does not increase. The role that endothelial cell plasticity plays in altering vascular tone and renal function, especially in response to changes in dietary salt intake, should be examined further in chronic kidney disease.
Collapse
Affiliation(s)
- Paul W Sanders
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, 642 Lyons-Harrison Research Building, 1530 Third Avenue South, Birmingham, AL 35294-0007, USA.
| |
Collapse
|
48
|
Abstract
The TGF-beta family comprises many structurally related differentiation factors that act through a heteromeric receptor complex at the cell surface and an intracellular signal transducing Smad complex. The receptor complex consists of two type II and two type I transmembrane serine/threonine kinases. Upon phosphorylation by the receptors, Smad complexes translocate into the nucleus, where they cooperate with sequence-specific transcription factors to regulate gene expression. The vertebrate genome encodes many ligands, fewer type II and type I receptors, and only a few Smads. In contrast to the perceived simplicity of the signal transduction mechanism with few Smads, the cellular responses to TGF-beta ligands are complex and context dependent. This raises the question of how the specificity of the ligand-induced signaling is achieved. We review the molecular basis for the specificity and versatility of signaling by the many ligands through this conceptually simple signal transduction mechanism.
Collapse
Affiliation(s)
- Xin-Hua Feng
- Department of Molecular and Cellular Biology, Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | |
Collapse
|
49
|
Abstract
During the past 10 years, it has been firmly established that Smad pathways are central mediators of signals from the receptors for transforming growth factor β (TGF-β) superfamily members to the nucleus. However, growing biochemical and developmental evidence supports the notion that alternative, non-Smad pathways also participate in TGF-β signalling. Non-Smad signalling proteins have three general mechanisms by which they contribute to physiological responses to TGF-β: (1) non-Smad signalling pathways directly modify (e.g. phosphorylate) the Smads and thus modulate the activity of the central effectors; (2) Smads directly interact and modulate the activity of other signalling proteins (e.g. kinases), thus transmitting signals to other pathways; and (3) the TGF-β receptors directly interact with or phosphorylate non-Smad proteins, thus initiating parallel signalling that cooperates with the Smad pathway in eliciting physiological responses. Thus, non-Smad signal transducers under the control of TGF-β provide quantitative regulation of the signalling pathway, and serve as nodes for crosstalk with other major signalling pathways, such as tyrosine kinase, G-protein-coupled or cytokine receptors.
Collapse
Affiliation(s)
- Aristidis Moustakas
- Ludwig Institute for Cancer Research, Biomedical Center, Uppsala University, Box 595, SE 751 24 Uppsala, Sweden.
| | | |
Collapse
|
50
|
Wesselkamper SC, Case LM, Henning LN, Borchers MT, Tichelaar JW, Mason JM, Dragin N, Medvedovic M, Sartor MA, Tomlinson CR, Leikauf GD. Gene expression changes during the development of acute lung injury: role of transforming growth factor beta. Am J Respir Crit Care Med 2005; 172:1399-411. [PMID: 16100012 PMCID: PMC2718437 DOI: 10.1164/rccm.200502-286oc] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RATIONALE Acute lung injury can occur from multiple causes, resulting in high mortality. The pathophysiology of nickel-induced acute lung injury in mice is remarkably complex, and the molecular mechanisms are uncertain. OBJECTIVES To integrate molecular pathways and investigate the role of transforming growth factor beta (TGF-beta) in acute lung injury in mice. METHODS cDNA microarray analyses were used to identify lung gene expression changes after nickel exposure. MAPPFinder analysis of the microarray data was used to determine significantly altered molecular pathways. TGF-beta1 protein in bronchoalveolar lavage fluid, as well as the effect of inhibition of TGF-beta, was assessed in nickel-exposed mice. The effect of TGF-beta on surfactant-associated protein B (Sftpb) promoter activity was measured in mouse lung epithelial cells. MEASUREMENTS AND MAIN RESULTS Genes that decreased the most after nickel exposure play important roles in lung fluid absorption or surfactant and phospholipid synthesis, and genes that increased the most were involved in TGF-beta signaling. MAPPFinder analysis further established TGF-beta signaling to be significantly altered. TGF-beta-inducible genes involved in the regulation of extracellular matrix function and fibrinolysis were significantly increased after nickel exposure, and TGF-beta1 protein was also increased in the lavage fluid. Pharmacologic inhibition of TGF-beta attenuated nickel-induced protein in bronchoalveolar lavage. In addition, treatment with TGF-beta1 dose-dependently repressed Sftpb promoter activity in vitro, and a novel TGF-beta-responsive region in the Sftpb promoter was identified. CONCLUSIONS These data suggest that TGF-beta acts as a central mediator of acute lung injury through the alteration of several different molecular pathways.
Collapse
Affiliation(s)
- Scott C Wesselkamper
- Department of Environmental Health, P.O. Box 670056, University of Cincinnati, Cincinnati, OH 45267-0056, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|