1
|
Wicaksono IA, Destiarani W, Romadhon SF, Nugraha BAP, Yusuf M, Milanda T, Amalia R. Transmembrane Prostate Androgen-Induced Protein 1 Molecular Modeling and Refinement Using Coarse-Grained Molecular Dynamics. ACS OMEGA 2025; 10:2712-2724. [PMID: 39895701 PMCID: PMC11780462 DOI: 10.1021/acsomega.4c08451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025]
Abstract
Transmembrane prostate androgen-induced protein 1 (TMEPAI), a type 1b transmembrane protein, is highly expressed in many types of cancer and is involved in cancer signaling pathways. TMEPAI affects the TGF-β, androgen receptor, Wnt, and MAPK/ERK signaling pathways. Although TMEPAI interactions are known, information about their structure is limited. This study performed TMEPAI structure prediction via a computational approach with template-free modeling using multiple Web server and refining with coarse-grained molecular dynamics to improve the understanding of its characterization, mechanism, and interactions, followed by intensive server-based validation. As a result, the predicted TMEPAI isoform structure was validated for all parameters, and the trRosetta server provided the most reliable predicted structure. This research is expected to provide preliminary scientific information about the TMEPAI structure prediction and apply it to develop targeted cancer therapy drugs.
Collapse
Affiliation(s)
- Imam Adi Wicaksono
- Department
of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
- Laboratory
of Translational Pharmaceutical Research, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
| | - Wanda Destiarani
- Department
of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Bandung 45363, Indonesia
| | - Shidqi Fajri Romadhon
- Department
of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
- Laboratory
of Translational Pharmaceutical Research, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
| | - Bagas Adi Prasetya Nugraha
- Department
of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
- Laboratory
of Translational Pharmaceutical Research, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
| | - Muhammad Yusuf
- Department
of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Bandung 45363, Indonesia
- Research
Center for Molecular Biotechnology and Bioinformatics, Universitas Padjadjaran, Bandung 45363, Indonesia
| | - Tiana Milanda
- Department
of Biology Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
| | - Riezki Amalia
- Department
of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
- Laboratory
of Translational Pharmaceutical Research, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
- Center
of
Excellence in Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung 45363, Indonesia
| |
Collapse
|
2
|
Huang H, Sun R, Xu Y, Liu R, Chen Z. PMEPA1 promotes gastric cancer cell proliferation by regulating the ubiquitin-mediated degradation of 14-3-3σ and promoting cell cycle progression. Braz J Med Biol Res 2024; 57:e13985. [PMID: 39607204 DOI: 10.1590/1414-431x2024e13985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/08/2024] [Indexed: 11/29/2024] Open
Abstract
Gastric cancer (GC) remains a global health challenge due to its heterogeneity and diverse regional epidemiology. Treatment for advanced GC often requires chemotherapy, whose effects are closely associated with the cell cycle. This association highlights the critical need to understand cell cycle regulators that can influence the effectiveness of chemotherapy. Bioinformatics analyses were performed on transcriptome data from a hospital cohort and on a publicly available database. Flow cytometry was used for cell cycle analysis. The interaction of PMEPA1 with 14-3-3σ was confirmed by coimmunoprecipitation and immunofluorescence staining. Western blot analysis was performed following inhibition of protein synthesis and degradation to assess 14-3-3σ protein stability, while ubiquitination was evaluated after treatment with the proteasome inhibitor MG132. High PMEPA1 expression was detected in GC tissues and was correlated with poor prognosis. In vitro overexpression of PMEPA1 promoted GC cell proliferation, while knockdown of PMEPA1 inhibited cell proliferation and induced G2/M arrest. In vivo study showed that overexpressing PMEPA1 promoted tumor growth, while knocking down PMEPA1 inhibited tumor growth, as indicated by the level of the proliferation marker Ki67. 14-3-3σ was identified as a downstream target of PMEPA1. PMEPA1 binds to 14-3-3σ and promoted its degradation by facilitating its ubiquitination. Overexpression of PMEPA1 increased its interactions with TTC3 and 14-3-3σ, increased 14-3-3σ ubiquitination, and reduced 14-3-3σ stability, and the opposite effects were observed after PMEPA1 knockdown. PMEPA1 recruited TTC3, allowing the ubiquitination of 14-3-3σ and leading to its degradation, thus promoting cell cycle progression in GC.
Collapse
Affiliation(s)
- Heyuan Huang
- The Hunan Provincial Key Lab of Precision Diagnosis and Treatment for Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruizheng Sun
- The Hunan Provincial Key Lab of Precision Diagnosis and Treatment for Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Xu
- The Hunan Provincial Key Lab of Precision Diagnosis and Treatment for Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Renchao Liu
- The Hunan Provincial Key Lab of Precision Diagnosis and Treatment for Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zihua Chen
- The Hunan Provincial Key Lab of Precision Diagnosis and Treatment for Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Hao Y, Xie F, He J, Gu C, Zhao Y, Luo W, Song X, Shen J, Yu L, Han Z, He J. PLA inhibits TNF-α-induced PANoptosis of prostate cancer cells through metabolic reprogramming. Int J Biochem Cell Biol 2024; 169:106554. [PMID: 38408537 DOI: 10.1016/j.biocel.2024.106554] [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: 11/14/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 02/28/2024]
Abstract
Previous studies have shown that phenyllactic acid (alpha-Hydroxyhydrocinnamic acid, 2-Hydroxy-3-phenylpropionic acid, PLA), a type of organic acid metabolite, has excellent diagnostic efficacy when used to differentiate between prostate cancer, benign prostatic hyperplasia, and prostatitis. This research aims to explore the molecular mechanism by which PLA influences the PANoptosis of prostate cancer (PCa) cell lines. First, we found that PLA was detected in all prostate cancer cell lines (PC-3, PC-3 M, DU145, LNCAP). Further experiments showed that the addition of PLA to prostate cancer cells could promote ATP generation, enhance cysteine desulfurase (NFS1) expression, and reduce tumor necrosis factor alpha (TNF-α) levels, thereby inhibiting apoptosis in prostate cancer cells. Notably, overexpression of NFS1 can inhibit the binding of TNF-α to serpin mRNA binding protein 1 (SERBP1), suggesting that NFS1 competes with TNF-α for binding to SERBP1. Knockdown of SERBP1 significantly reduced the level of small ubiquity-related modifier (SUMO) modification of TNF-α. This suggests that NFS1 reduces the SUMO modification of TNF-α by competing with SERBP1, thereby reducing the expression and stability of TNF-α and ultimately inhibiting apoptosis in prostate cancer cell lines. In conclusion, PLA inhibits TNF-α induced panapoptosis of prostate cancer cells through metabolic reprogramming, providing a new idea for targeted treatment of prostate cancer.
Collapse
Affiliation(s)
- Yinghui Hao
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Fangmei Xie
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jieyi He
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Chenqiong Gu
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Ying Zhao
- Central Laboratory of Panyu Central Hospital, Guangzhou, China
| | - Wenfeng Luo
- Central Laboratory of Panyu Central Hospital, Guangzhou, China
| | - Xiaoyu Song
- Central Laboratory of Panyu Central Hospital, Guangzhou, China
| | - Jian Shen
- Central Laboratory of Panyu Central Hospital, Guangzhou, China
| | - Li Yu
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China.
| | - Zeping Han
- Central Laboratory of Panyu Central Hospital, Guangzhou, China.
| | - Jinhua He
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China; Central Laboratory of Panyu Central Hospital, Guangzhou, China; Rehabilitation Medicine Institute of Panyu District, Guangzhou, China.
| |
Collapse
|
4
|
Zhu Q, Wang Y, Liu Y, Yang X, Shuai Z. Prostate transmembrane androgen inducible protein 1 (PMEPA1): regulation and clinical implications. Front Oncol 2023; 13:1298660. [PMID: 38173834 PMCID: PMC10761476 DOI: 10.3389/fonc.2023.1298660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Prostate transmembrane androgen inducible protein 1 (PMEPA1) can promote or inhibit prostate cancer cell growth based on the cancer cell response to the androgen receptor (AR). Further, it can be upregulated by transforming growth factor (TGF), which downregulates transforming growth factor-β (TGF-β) signaling by interfering with R-Smad phosphorylation to facilitate TGF-β receptor degradation. Studies have indicated the increased expression of PMEPA1 in some solid tumors and its functioning as a regulator of multiple signaling pathways. This review highlights the multiple potential signaling pathways associated with PMEPA1 and the role of the PMEPA1 gene in regulating prognosis, including transcriptional regulation and epithelial mesenchymal transition (EMT). Moreover, the relevant implications in and outside tumors, for example, as a biomarker and its potential functions in lysosomes have also been discussed.
Collapse
Affiliation(s)
- Qicui Zhu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yue Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yaqian Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoke Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zongwen Shuai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| |
Collapse
|
5
|
Li Y, Zhang Y, Li L, Zhang M, Song N, Zhao Q, Liu Z, Diao A. TMEPAI promotes degradation of the NF-κB signaling pathway inhibitory protein IκBα and contributes to tumorigenesis. Int J Biol Macromol 2023; 235:123859. [PMID: 36868334 DOI: 10.1016/j.ijbiomac.2023.123859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023]
Abstract
The transmembrane prostate androgen-induced protein (TMEPAI) is known to be highly expressed in various types of cancer and promoted oncogenic abilities. However, the mechanisms whereby TMEPAI facilitates tumorigenesis are not fully understood. Here we reported that expression of TMEPAI activated the NF-κB signaling. TMEPAI showed direct interaction with NF-κB pathway inhibitory protein IκBα. Though ubiquitin ligase Nedd4 (neural precursor cell expressed, developmentally down-regulated 4) did not interact with IκBα directly, TMEPAI recruited Nedd4 for ubiquitination of IκBα, leading to IκBα degradation through the proteasomal and lysosomal pathway, and promoted activation of NF-κB signaling. Further study indicated NF-κB signaling is involved in TMEPAI-induced cell proliferation and tumor growth in immune deficient mice. This finding helps to further understand the mechanism of TMEPAI on tumorigenesis and suggests TMEPAI is potential target for cancer treatment.
Collapse
Affiliation(s)
- Yuyin Li
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, State Key Laboratory of Food Nutrition and Safety, Tianjin 300457, China.
| | - Yaxin Zhang
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, State Key Laboratory of Food Nutrition and Safety, Tianjin 300457, China
| | - Lu Li
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, State Key Laboratory of Food Nutrition and Safety, Tianjin 300457, China
| | - Mei Zhang
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, State Key Laboratory of Food Nutrition and Safety, Tianjin 300457, China
| | - Ning Song
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, State Key Laboratory of Food Nutrition and Safety, Tianjin 300457, China
| | - Qing Zhao
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, State Key Laboratory of Food Nutrition and Safety, Tianjin 300457, China
| | - Zhenxing Liu
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, State Key Laboratory of Food Nutrition and Safety, Tianjin 300457, China
| | - Aipo Diao
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, State Key Laboratory of Food Nutrition and Safety, Tianjin 300457, China.
| |
Collapse
|
6
|
Yu ZL, Liu J, Ning ZK, Tian HK, Wu X, Huang YF, Wu ZC, Zong Z, Zhou TC. The TGF-β/Smad 2/3 signaling pathway is involved in Musashi2-induced invasion and metastasis of colorectal cancer. Mol Carcinog 2023; 62:261-276. [PMID: 36345938 DOI: 10.1002/mc.23484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/08/2022] [Accepted: 06/20/2022] [Indexed: 11/11/2022]
Abstract
To identify Musashi2 as an effective biomarker regulated by the TGF-β/Smad2/3 signaling pathway for the precise diagnosis and treatment of colorectal cancer (CRC) through bioinformatic tools and experimental verification. The Cancer Genome Atlas, Timer, and Kaplan-Meier analyses were performed to clarify the expression of Musashi2 and its influence on the prognosis of CRC. Transforming growth factor beta 1 (TGF-β1) was used to activate the TGF-β/Smad2/3 signaling pathway to identify whether it could regulate the expression and function of Musashi2. Western blot analysis and quantitative PCR analyses were conducted to verify the expression of Musashi2. Cell counting kit-8 (CCK8), EdU, wound healing, and Transwell assays were conducted to reveal the role of Musashi2 in the proliferation, migration, and invasion of CRC. Musashi2 was upregulated in CRC and promoted proliferation and metastasis. TGF-β1 increased the expression of Musashi2, while the antagonist inducer of type II TGF-β receptor degradation-1 (ITD-1) decreased the expression. CCK8 and EdU assays demonstrated that inhibition of Musashi2 or use of ITD-1 lowered proliferation ability. The Transwell and wound healing assays showed that the migration and invasion abilities of CRC cells could be regulated by Musashi2. The above functions could be enhanced by TGF-β1 by activating the TGF-β/Smad2/3 signaling pathway and reversed by ITD-1. A positive correlation was found between Musashi2 and the TGF-β/Smad2/3 signaling pathway. TGF-β1 activates the TGF-β/Smad2/3 pathway to stimulate the expression of Musashi2, which promotes the progression of CRC. Musashi2 might become a target gene for the development of new antitumor drugs.
Collapse
Affiliation(s)
- Zhong Lin Yu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiang Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhi Kun Ning
- Department of Day Ward, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hua Kai Tian
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xun Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ying Feng Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zi Chun Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tai Cheng Zhou
- Department of Gastroenterological Surgery and Hernia Center, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Shabestari AN, Tamehri Zadeh SS, Zahmatkesh P, Baghdadabad LZ, Mirzaei A, Mashhadi R, Mesbah G, Khajavi A, Akbarzadehmoallemkolaei M, Khoshchehreh M, Rahimnia R, Kazem Aghamir SM. The Impact of Conventional Smoking versus Electronic Cigarette on the Expression of VEGF, PEMPA1, and PTEN in Rat Prostate. Prostate Int 2022. [DOI: 10.1016/j.prnil.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
8
|
Zhang C, Wang S, Casal Moura M, Yi ES, Bowen AJ, Specks U, Warrington KJ, Bayan SL, Ekbom DC, Luo F, Edell ES, Kasperbauer JL, Vassallo R. RNA Sequencing of Idiopathic Subglottic Stenosis Tissues Uncovers Putative Profibrotic Mechanisms and Identifies a Prognostic Biomarker. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1506-1530. [PMID: 35948078 DOI: 10.1016/j.ajpath.2022.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/30/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023]
Abstract
Idiopathic subglottic stenosis (iSGS) is a localized airway disease that almost exclusively affects females. Understanding the molecular mechanisms involved may provide insights leading to therapeutic interventions. Next-generation sequencing was performed on tissue sections from patients with iSGS (n = 22), antineutrophil cytoplasmic antibody-associated vasculitis (AAV; n = 5), and matched controls (n = 9) to explore candidate genes and mechanisms of disease. Gene expression changes were validated, and selected markers were identified by immunofluorescence staining. Epithelial-mesenchymal transition (EMT) and leukocyte extravasation pathways were the biological mechanisms most relevant to iSGS pathogenesis. Alternatively activated macrophages (M2) were abundant in the subepithelium and perisubmucosal glands of the airway in iSGS and AAV. Increased expression of the mesenchymal marker S100A4 and decreased expression of the epithelial marker epithelial cell adhesion molecule (EPCAM) further supported a role for EMT, but to different extents, in iSGS and antineutrophil cytoplasmic antibody-associated subglottic stenosis. In patients with iSGS, high expression of prostate transmembrane protein, androgen induced 1 (PMEPA1), an EMT regulator, was associated with a shorter recurrence interval (25 versus 116 months: hazard ratio = 4.16; P = 0.041; 95% CI, 1.056-15.60). Thus, EMT is a key pathogenetic mechanism of subglottic stenosis in iSGS and AAV. M2 macrophages contribute to the pathogenesis of both diseases, suggesting a shared profibrotic mechanism, and PMEPA1 may be a biomarker for predicting disease recurrence in iSGS.
Collapse
Affiliation(s)
- Chujie Zhang
- Division of Pulmonary and Critical Care Medicine and Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota; Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Shaohua Wang
- Division of Pulmonary and Critical Care Medicine and Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Marta Casal Moura
- Division of Pulmonary and Critical Care Medicine and Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Eunhee S Yi
- Departments of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Andrew J Bowen
- Otorhinolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine and Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | | | - Semirra L Bayan
- Otorhinolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Dale C Ekbom
- Otorhinolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Fengming Luo
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Eric S Edell
- Division of Pulmonary and Critical Care Medicine and Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Jan L Kasperbauer
- Otorhinolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Robert Vassallo
- Division of Pulmonary and Critical Care Medicine and Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
9
|
The Synergistic Cooperation between TGF-β and Hypoxia in Cancer and Fibrosis. Biomolecules 2022; 12:biom12050635. [PMID: 35625561 PMCID: PMC9138354 DOI: 10.3390/biom12050635] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 03/10/2022] [Accepted: 04/14/2022] [Indexed: 12/24/2022] Open
Abstract
Transforming growth factor β (TGF-β) is a multifunctional cytokine regulating homeostasis and immune responses in adult animals and humans. Aberrant and overactive TGF-β signaling promotes cancer initiation and fibrosis through epithelial–mesenchymal transition (EMT), as well as the invasion and metastatic growth of cancer cells. TGF-β is a key factor that is active during hypoxic conditions in cancer and is thereby capable of contributing to angiogenesis in various types of cancer. Another potent role of TGF-β is suppressing immune responses in cancer patients. The strong tumor-promoting effects of TGF-β and its profibrotic effects make it a focus for the development of novel therapeutic strategies against cancer and fibrosis as well as an attractive drug target in combination with immune regulatory checkpoint inhibitors. TGF-β belongs to a family of cytokines that exert their function through signaling via serine/threonine kinase transmembrane receptors to intracellular Smad proteins via the canonical pathway and in combination with co-regulators such as the adaptor protein and E3 ubiquitin ligases TRAF4 and TRAF6 to promote non-canonical pathways. Finally, the outcome of gene transcription initiated by TGF-β is context-dependent and controlled by signals exerted by other growth factors such as EGF and Wnt. Here, we discuss the synergistic cooperation between TGF-β and hypoxia in development, fibrosis and cancer.
Collapse
|
10
|
Wang CI, Chu PM, Chen YL, Lin YH, Chen CY. Chemotherapeutic Drug-Regulated Cytokines Might Influence Therapeutic Efficacy in HCC. Int J Mol Sci 2021; 22:ijms222413627. [PMID: 34948424 PMCID: PMC8707970 DOI: 10.3390/ijms222413627] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/04/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC), the most common type of liver cancer, is the second leading cause of cancer-related mortality worldwide. Processes involved in HCC progression and development, including cell transformation, proliferation, metastasis, and angiogenesis, are inflammation-associated carcinogenic processes because most cases of HCC develop from chronic liver damage and inflammation. Inflammation has been demonstrated to be a crucial factor inducing tumor development in various cancers, including HCC. Cytokines play critical roles in inflammation to accelerate tumor invasion and metastasis by mediating the migration of immune cells into damaged tissues in response to proinflammatory stimuli. Currently, surgical resection followed by chemotherapy is the most common curative therapeutic regimen for HCC. However, after chemotherapy, drug resistance is clearly observed, and cytokine secretion is dysregulated. Various chemotherapeutic agents, including cisplatin, etoposide, and 5-fluorouracil, demonstrate even lower efficacy in HCC than in other cancers. Tumor resistance to chemotherapeutic drugs is the key limitation of curative treatment and is responsible for treatment failure and recurrence, thus limiting the ability to treat patients with advanced HCC. Therefore, the capability to counteract drug resistance would be a major clinical advancement. In this review, we provide an overview of links between chemotherapeutic agents and inflammatory cytokine secretion in HCC. These links might provide insight into overcoming inflammatory reactions and cytokine secretion, ultimately counteracting chemotherapeutic resistance.
Collapse
Affiliation(s)
- Chun-I Wang
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan;
| | - Pei-Ming Chu
- Department of Anatomy, School of Medicine, China Medical University, Taichung 404, Taiwan;
| | - Yi-Li Chen
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Yang-Hsiang Lin
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan;
| | - Cheng-Yi Chen
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
- Correspondence: ; Tel./Fax: +886-6-2353535 (ext. 5329)
| |
Collapse
|
11
|
Qiu D, Hu J, Hu J, Yu A, Othmane B, He T, Ding J, Cheng X, Ren W, Tan X, Yu Q, Chen J, Zu X. PMEPA1 Is a Prognostic Biomarker That Correlates With Cell Malignancy and the Tumor Microenvironment in Bladder Cancer. Front Immunol 2021; 12:705086. [PMID: 34777336 PMCID: PMC8582246 DOI: 10.3389/fimmu.2021.705086] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 10/05/2021] [Indexed: 01/02/2023] Open
Abstract
Prostate transmembrane protein androgen induced 1 (PMEPA1) has been reported to promote cancer progression, but the potential role of PMEPA1 in bladder cancer (BLCA) remains elusive. We assess the role of PMEPA1 in BLCA, via a publicly available database and in vitro study. PMEPA1 was identified from 107 differentially expressed genes (DEGs) to have prognostic value. GO, KEGG, and GSEA analysis indicated that PMEPA1 was involved in cancer progression and the tumor microenvironment (TME). Then bioinformatical analysis in TCGA, GEO, TIMER, and TISIDB show a positive correlation with the inflammation and infiltration levels of three tumor-infiltrating immune cells (TAMs, CAFs, and MDSCs) and immune/stromal scores in TME. Moreover, in vitro study revealed that PMEPA1 promotes bladder cancer cell malignancy. Immunohistochemistry and survival analysis shed light on PMEPA1 potential to be a novel biomarker in predicting tumor progression and prognosis. At last, we also analyzed the role of PMEPA1 in predicting the molecular subtype and the response to several treatment options in BLCA. We found that PMEPA1 may be a novel potential biomarker to predict the progression, prognosis, and molecular subtype of BLCA.
Collapse
Affiliation(s)
- Dongxu Qiu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,Department of Urology, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Jiao Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Anze Yu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,Immunobiology & Transplant Science Center, Houston Methodist Research Institute, Texas Medical Center, Houston, TX, United States
| | - Belaydi Othmane
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Tongchen He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Ding
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Xu Cheng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,Institute for Infection Prevention and Hospital Epidemiology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Wenbiao Ren
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, United States
| | - Xiyan Tan
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Qiaoyan Yu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
TGF-β Increases MFGE8 Production in Myeloid-Derived Suppressor Cells to Promote B16F10 Melanoma Metastasis. Biomedicines 2021; 9:biomedicines9080896. [PMID: 34440100 PMCID: PMC8389657 DOI: 10.3390/biomedicines9080896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/12/2021] [Accepted: 07/23/2021] [Indexed: 01/11/2023] Open
Abstract
There is growing evidence that myeloid-derived suppressor cells (MDSCs) are directly involved in all stages leading to metastasis. Many mechanisms for this effect have been proposed, but mechanisms of coregulation between tumor cells and MDSCs remain poorly understood. In this study, we demonstrate that MDSCs are a source of milk fat globule-epidermal growth factor (EGF) factor 8 (MFGE8), which is known to be involved in tumor metastasis. Interestingly, TGF-β, an abundant cytokine in the tumor microenvironment (TME), increased MFGE8 production by MDSCs. In addition, co-culturing MDSCs with B16F10 melanoma cells increased B16F10 cell migration, while MFGE8 neutralization decreased their migration. Taken together, these findings suggest that MFGE8 is an important effector molecule through which MDSCs promote tumor metastasis, and the TME positively regulates MFGE8 production by MDSCs through TGF-β.
Collapse
|
13
|
Sharad S, Dobi A, Srivastava S, Srinivasan A, Li H. PMEPA1 Gene Isoforms: A Potential Biomarker and Therapeutic Target in Prostate Cancer. Biomolecules 2020; 10:biom10091221. [PMID: 32842649 PMCID: PMC7565192 DOI: 10.3390/biom10091221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/26/2022] Open
Abstract
The identification of prostate transmembrane protein androgen induced 1 (PMEPA1), an androgen responsive gene, came initially from the studies of androgen regulatory gene networks in prostate cancer. It was soon followed by the documentation of the expression and functional analysis of transmembrane prostate androgen-induced protein (TMEPAI)/PMEPA1 in other solid tumors including renal, colon, breast, lung, and ovarian cancers. Further elucidation of PMEPA1 gene expression and sequence analysis revealed the presence of five isoforms with distinct extracellular domains (isoforms a, b, c, d, and e). Notably, the predicted amino acid sequences of PMEPA1 isoforms show differences at the N-termini, a conserved membrane spanning and cytoplasmic domains. PMEPA1 serves as an essential regulator of multiple signaling pathways including androgen and TGF-β signaling in solid tumors. Structure-function studies indicate that specific motifs present in the cytoplasmic domain (PY, SIM, SH3, and WW binding domains) are utilized to mediate isoform-specific functions through interactions with other proteins. The understanding of the “division of labor” paradigm exhibited by PMEPA1 isoforms further expands our knowledge of gene’s multiple functions in tumorigenesis. In this review, we aim to summarize the most recent advances in understanding of PMEPA1 isoform-specific functions and their associations with prostate cancer progression, highlighting the potentials as biomarker and therapeutic target in prostate cancer.
Collapse
Affiliation(s)
- Shashwat Sharad
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD 20817, USA; (A.D.); (S.S.); (A.S.)
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
- Correspondence: (S.S.); (H.L.); Tel.: +1-240-694-4931 (S.S.); +1-240-694-4944 (H.L.)
| | - Albert Dobi
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD 20817, USA; (A.D.); (S.S.); (A.S.)
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Shiv Srivastava
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD 20817, USA; (A.D.); (S.S.); (A.S.)
| | - Alagarsamy Srinivasan
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD 20817, USA; (A.D.); (S.S.); (A.S.)
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Hua Li
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD 20817, USA; (A.D.); (S.S.); (A.S.)
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
- Correspondence: (S.S.); (H.L.); Tel.: +1-240-694-4931 (S.S.); +1-240-694-4944 (H.L.)
| |
Collapse
|