1
|
Khan A, Alzahrani HA, Felemban SG, Algarni AS, Alenezi ABS, Kamal M, Rehman ZU, Asdaq SMB, Ahmed N, Alharbi BM, Alanazi BS, Imran M. Exploring TGF-β signaling in benign prostatic hyperplasia: from cellular senescence to fibrosis and therapeutic implications. Biogerontology 2025; 26:79. [PMID: 40159577 DOI: 10.1007/s10522-025-10226-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
As men get older, they often develop benign prostatic hyperplasia (BPH), an enlarged prostate that is not cancerous or dangerous. Although the etiology of BPH is unknown, increasing evidence indicates that the TGF-β signaling pathway might be a key player in its pathogenesis. TGF-β is a pleiotropic cytokine involved in proliferation, differentiation, and extracellular matrix re-modeling, which are all dysregulated in BPH. Cellular senescence is primarily initiated by TGF-β--induced, irreversible growth arrest and usually limits the prostate gland's hyperplastic growth. Moreover, senescent cells generate a Senescence-Associated Secretory Phenotype (SASP), which consists of numerous proinflammatory and profibrotic factors that can worsen disease ontogeny. In addition, TGF-β is among the most fibrogenic factors. At the same time, fibrosis involves a massive accumulation of extracellular matrix proteins, which can increase tissue stiffness and a loss of normal organ functions. TGF-β-mediated fibrosis in BPH changes the mechanical properties of the prostate and surrounding tissues to contribute to lower urinary tract symptoms. This review discusses the complicated molecular signaling of TGF-β underlying changes in cellular senescence and fibrosis during BPH concerning its therapeutic potential.
Collapse
Affiliation(s)
- Abida Khan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, 73213, Saudi Arabia
| | - Hayat Ali Alzahrani
- Medical Laboratory Technology Department, College of Medical Applied Science, Northern Border University, Arar, Saudi Arabia
| | - Shatha Ghazi Felemban
- Medical Laboratory Sciences Department, Fakeeh College for Medical Sciences, 21461, Jeddah, Saudi Arabia
| | - Alanood Saeed Algarni
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Zia Ur Rehman
- Health Research Centre, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jazan University, P.O. Box 114, Jazan, 45142, Kingdom of Saudi Arabia
| | | | - Naveed Ahmed
- Department of Assistance Medical Sciences, Applied College, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Bashayer Mohammed Alharbi
- Department of Pharmacy, Johns Hopkins Aramco Healthcare, P.O. Box 10352, 31311, Dhahran, Eastern Province, Saudi Arabia
| | - Bander Sharqi Alanazi
- Department of Nursing Administration, Northern Area Armed Forces Hospital, 31991, Hafer AlBaten, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, 73213, Saudi Arabia.
| |
Collapse
|
2
|
Hu S, Gao X, Zhu Y, Shi F, Huang L. PMEPA1 Binds NEDD4L to Inhibit the Malignant Progression of Multiple Myeloma by Inactivating Wnt/β-Catenin Signaling. Cell Biochem Biophys 2025:10.1007/s12013-025-01674-w. [PMID: 40035958 DOI: 10.1007/s12013-025-01674-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2025] [Indexed: 03/06/2025]
Abstract
Multiple myeloma (MM) is an incurable hematological malignancy with increasing prevalence. Prostate transmembrane androgen inducible protein 1 (PMEPA1) is positively associated with overall survival in MM patients, but the exact functions and mechanisms of PMEPA1 in MM have yet to be elucidated. PMEPA1 and neural precursor cell-expressed developmentally downregulated gene 4L (NEDD4L) levels in MM cells were examined. In RPMI-8226 cells with PMEPA1 overexpression or/and NEDD4L knockdown, cell proliferation, cycle distribution and apoptosis were evaluated with the application of CCK-8, EDU staining and flow cytometry. The BioGrid website and HDOCK SERVER were applied for predicting the binding between PMEPA1 and NEDD4L, which was checked by co-immunoprecipitation. Besides, the levels of proteins associated with proliferation (Ki67 and PCNA), apoptosis (Bcl-2, Bax and cleaved caspase3) and Wnt/β-catenin signaling (β-catenin, c-Myc and cyclin D1) was detected with immunoblotting. Finally, LiCl, an activator of Wnt/β-catenin pathway, was employed to treat RPMI-8226 cells to analyze the proliferation, cycle distribution and apoptosis of MM cells. As a result, PMEPA1 and NEDD4L were expressed at low levels in MM cells. PMEPA1 upregulation repressed proliferation induced cycle arrest and facilitated apoptosis of MM cells. Moreover, PMEPA1 bound to NEDD4L and upregulated NEDD4L expression in RPMI-8226 cells. Functionally, NEDD4L knockdown attenuated the influences of PMEPA1 overexpression on the proliferation, cycle distribution and apoptosis of RPMI-8226 cells. Additionally, PMEPA1 notably downregulated β-catenin, c-Myc and cyclin D1 expression in RPMI-8226 cells, which was abrogated by NEDD4L silencing. Further adding LiCl in RPMI-8226 cells led to the enhanced malignant biological behaviors. Collectively, PMEPA1 damaged MM progression through binding NEDD4L to inactivate Wnt/β-catenin signaling, which may be helpful to develop promising targets for MM treatment.
Collapse
Affiliation(s)
- Shanshan Hu
- Department of Hematology, Jinhua People's Hospital, Jinhua City, Zhejiang Province, 321000, China
| | - Xinfang Gao
- Department of Hematology, Jinhua People's Hospital, Jinhua City, Zhejiang Province, 321000, China
| | - Yan Zhu
- Department of Hematology, Jinhua People's Hospital, Jinhua City, Zhejiang Province, 321000, China
| | - Fangjing Shi
- Department of Hematology, Jinhua People's Hospital, Jinhua City, Zhejiang Province, 321000, China
| | - Li Huang
- Department of Hematology, Jinhua People's Hospital, Jinhua City, Zhejiang Province, 321000, China.
| |
Collapse
|
3
|
Song J, Liu Z, Yang F, Zhang T, Pan Z. Hypomethylation of the low-density lipoprotein receptor class A domain containing 4 gene in rheumatoid arthritis. Clin Immunol 2025; 272:110441. [PMID: 39870147 DOI: 10.1016/j.clim.2025.110441] [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: 11/16/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory disease linked to epigenetic changes, particularly DNA methylation. While LDLRAD4 has been implicated in RA through GWAS, its role in RA via methylation remains unclear. OBJECTIVES To investigate LDLRAD4 methylation patterns in RA and evaluate its potential as a diagnostic and inflammatory biomarker. METHODS We assessed DNA methylation at specific CpG sites within LDLRAD4 in 150 RA patients and 150 healthy controls. Clinical data, including disease duration and inflammatory markers, were collected. RESULTS RA patients showed significant hypomethylation of LDLRAD4, especially in the LDLRAD4-43 and LDLRAD4-44 regions. ROC analysis yielded an AUC of 0.841, indicating strong diagnostic potential. Methylation levels correlated negatively with ESR, CRP and DAS28 in the RF+/CCP- subgroup. CONCLUSIONS LDLRAD4 DNA present hypomethylation in rheumatoid arthritis, and methylation levels are correlated with inflammatory indicator, possibly via TGF-β signaling. Further research is needed to explore its therapeutic potential.
Collapse
Affiliation(s)
- Jingjing Song
- Department of Rheumatology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Zhen Liu
- Department of Rheumatology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Fan Yang
- Department of Rheumatology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Ting Zhang
- Department of Rheumatology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Zhenglun Pan
- Department of Rheumatology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China.
| |
Collapse
|
4
|
Ishiguro H, Ushiki T, Honda A, Yoshimatsu Y, Ohashi R, Okuda S, Kawasaki A, Cho K, Tamura S, Suwabe T, Katagiri T, Ling Y, Iijima A, Mikami T, Kitagawa H, Uemura A, Sango K, Masuko M, Igarashi M, Sone H. Reduced chondroitin sulfate content prevents diabetic neuropathy through transforming growth factor-β signaling suppression. iScience 2024; 27:109528. [PMID: 38595797 PMCID: PMC11002665 DOI: 10.1016/j.isci.2024.109528] [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: 07/31/2023] [Revised: 11/08/2023] [Accepted: 03/15/2024] [Indexed: 04/11/2024] Open
Abstract
Diabetic neuropathy (DN) is a major complication of diabetes mellitus. Chondroitin sulfate (CS) is one of the most important extracellular matrix components and is known to interact with various diffusible factors; however, its role in DN pathology has not been examined. Therefore, we generated CSGalNAc-T1 knockout (T1KO) mice, in which CS levels were reduced. We demonstrated that diabetic T1KO mice were much more resistant to DN than diabetic wild-type (WT) mice. We also found that interactions between pericytes and vascular endothelial cells were more stable in T1KO mice. Among the RNA-seq results, we focused on the transforming growth factor β signaling pathway and found that the phosphorylation of Smad2/3 was less upregulated in T1KO mice than in WT mice under hyperglycemic conditions. Taken together, a reduction in CS level attenuates DN progression, indicating that CS is an important factor in DN pathogenesis.
Collapse
Affiliation(s)
- Hajime Ishiguro
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| | - Takashi Ushiki
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
- Division of Hematology and Oncology, Graduate School of Health Sciences, Niigata University, Niigata, Japan
- Departments of Transfusion Medicine, Cell Therapy and Regenerative Medicine, Medical and Dental Hospital, Niigata University, Niigata, Japan
| | - Atsuko Honda
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Center for Research Promotion, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yasuhiro Yoshimatsu
- Division of Pharmacology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Riuko Ohashi
- Divisions of Molecular and Diagnostic Pathology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kaori Cho
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| | - Suguru Tamura
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| | - Tatsuya Suwabe
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| | - Takayuki Katagiri
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| | - Yiwei Ling
- Division of Bioinformatics, Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Atsuhiko Iijima
- Neurophysiology & Biomedical Engineering Lab, Interdisciplinary Program of Biomedical Engineering, Assistive Technology and Art and Sports Sciences, Faculty of Engineering, Niigata University Niigata, Niigata, Japan
| | - Tadahisa Mikami
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kazunori Sango
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masayoshi Masuko
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
- Hematopoietic Cell Transplantation Niigata University Medical and Dental Hospital, , Niigata University, Niigata, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hirohito Sone
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| |
Collapse
|
5
|
Aßfalg M, Güner G, Müller SA, Breimann S, Langosch D, Muhle-Goll C, Frishman D, Steiner H, Lichtenthaler SF. Cleavage efficiency of the intramembrane protease γ-secretase is reduced by the palmitoylation of a substrate's transmembrane domain. FASEB J 2024; 38:e23442. [PMID: 38275103 DOI: 10.1096/fj.202302152r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/20/2023] [Accepted: 01/09/2024] [Indexed: 01/27/2024]
Abstract
The intramembrane protease γ-secretase has broad physiological functions, but also contributes to Notch-dependent tumors and Alzheimer's disease. While γ-secretase cleaves numerous membrane proteins, only few nonsubstrates are known. Thus, a fundamental open question is how γ-secretase distinguishes substrates from nonsubstrates and whether sequence-based features or post-translational modifications of membrane proteins contribute to substrate recognition. Using mass spectrometry-based proteomics, we identified several type I membrane proteins with short ectodomains that were inefficiently or not cleaved by γ-secretase, including 'pituitary tumor-transforming gene 1-interacting protein' (PTTG1IP). To analyze the mechanism preventing cleavage of these putative nonsubstrates, we used the validated substrate FN14 as a backbone and replaced its transmembrane domain (TMD), where γ-cleavage occurs, with the one of nonsubstrates. Surprisingly, some nonsubstrate TMDs were efficiently cleaved in the FN14 backbone, demonstrating that a cleavable TMD is necessary, but not sufficient for cleavage by γ-secretase. Cleavage efficiencies varied by up to 200-fold. Other TMDs, including that of PTTG1IP, were still barely cleaved within the FN14 backbone. Pharmacological and mutational experiments revealed that the PTTG1IP TMD is palmitoylated, which prevented cleavage by γ-secretase. We conclude that the TMD sequence of a membrane protein and its palmitoylation can be key factors determining substrate recognition and cleavage efficiency by γ-secretase.
Collapse
Affiliation(s)
- Marlene Aßfalg
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Gökhan Güner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stephan Breimann
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Bioinformatics, School of Life Sciences, Technical University of Munich, Freising, Germany
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, LMU Munich, Munich, Germany
| | | | - Claudia Muhle-Goll
- Institute for Biological Interfaces 4, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Dmitrij Frishman
- Department of Bioinformatics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Harald Steiner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
6
|
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
|
7
|
Wen F, Yang S, Cai W, Zhao M, Qin L, Jiao Z. Exploring the role of PMEPA1 in gastric cancer. Mol Cell Probes 2023; 72:101931. [PMID: 37683830 DOI: 10.1016/j.mcp.2023.101931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
Although there are several treatments available for gastric cancer (GC), the prognosis of the disease is still poor due to many factors, such as late diagnosis and tumor heterogeneity. To identify potential therapeutic targets, bioinformatics techniques and clinical sample validation were employed and prostate transmembrane protein androgen induced 1 (PMEPA1) was selected for further study. In the present study, we found that elevated PMEPA1 expression correlates with a worse prognosis and weaker anti-tumor immunity in GC patients. Moreover, our study showed that PMEPA1 not only influences cell proliferation, clone formation, invasion, and migration in vitro, but also plays an important role in GC progression in vivo. Mechanically, PMEPA1 exerts its oncogenic effects through activating the Wnt/β-catenin signaling pathway. Therefore, PMEPA1 is a potential target for treating GC effectively.
Collapse
Affiliation(s)
- Fei Wen
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Shangyu Yang
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - WeiWen Cai
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Mengyuan Zhao
- Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Long Qin
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| | - Zuoyi Jiao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| |
Collapse
|
8
|
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
|
9
|
Wardhani BWK, Louisa M, Watanabe Y, Setiabudy R, Kato M. TGF-β-Induced TMEPAI Promotes Epithelial-Mesenchymal Transition in Doxorubicin-Treated Triple-Negative Breast Cancer Cells via SMAD3 and PI3K/AKT Pathway Alteration. BREAST CANCER-TARGETS AND THERAPY 2021; 13:529-538. [PMID: 34584450 PMCID: PMC8464328 DOI: 10.2147/bctt.s325429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/03/2021] [Indexed: 12/27/2022]
Abstract
Introduction Epithelial–mesenchymal transition (EMT) and overexpression of drug efflux transporters have been reported to cause doxorubicin resistance. Our previous study indicated that TMEPAI (transmembrane prostate androgen-induced protein) attenuated doxorubicin sensitivity in triple-negative breast cancer cells. However, how TMEPAI contributes to doxorubicin resistance in TNBC remains unclear. Thus, the present study aimed to elucidate the mechanism of TMEPAI in doxorubicin resistance in triple-negative breast cancer cells. Methods We used BT549, triple-negative cells wild type (WT), and BT549 TMEPAI knock-out. Both cells were treated with TGF-β 2 ng/mL for 24 hours, followed by TGF-β 2 ng/mL and doxorubicin 12.9 nM for another 24 hours. Afterward, the cells were harvested and counted. Cells were further lysed and used for RT-PCR and Western blot analysis. We determined the expression levels of proliferation, apoptosis, EMT markers, and drug efflux transporters. Additionally, we investigated the expressions of PI3K as well as SMAD3 and AKT phosphorylation. Results TNBC cells were shown to be less sensitive to doxorubicin in the presence of TMEPAI. TMEPAI was shown to alleviate the mRNA expressions of apoptosis markers: Bax, Bcl2, Caspase-3, and Caspase-9. Our results indicated that the presence of TMEPAI greatly amplifies EMT and increases drug efflux transporter expressions after doxorubicin treatment. Furthermore, our findings demonstrated that TMEPAI reduced the action of doxorubicin in inhibiting SMAD3 phosphorylation. TMEPAI was also shown to modify the effect of doxorubicin by reducing PI3K expressions and Akt phosphorylation in triple-negative breast cancer cells. Conclusion Our findings indicate that TMEPAI promotes EMT and drug efflux transporters at least in part by shifting doxorubicin action from SMAD3 phosphorylation reduction to PI3K/AKT inhibition in triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Bantari W K Wardhani
- Biomedical Sciences, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Department of Pharmacology, Faculty of Military Pharmacy, Indonesia Defense University, West Java, Indonesia
| | - Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Yukihide Watanabe
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Rianto Setiabudy
- Department of Pharmacology and Therapeutics, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Mitsuyasu Kato
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
10
|
Song M, Zhou B, Li B, Tian L. PMEPA1 Stimulates the Proliferation, Colony Formation of Pancreatic Cancer Cells via the MAPK Signaling Pathway. Am J Med Sci 2021; 362:291-296. [PMID: 33857498 DOI: 10.1016/j.amjms.2021.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/30/2020] [Accepted: 04/07/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Prostate transmembrane protein androgen-induced 1 (PMEPA1) is reportedly highly expressed in pancreatic cancer (PC). However, its biological role and associated mechanisms have not been addressed in PC progression. METHODS PMEPA1 mRNA expression and survival outcome of PC patients were evaluated via the GEPIA website. Lentiviral-mediated shRNA knockdown and ectopic expression of PMEPA1 were implemented in the pancreatic cancer cell line PANC1 cells. CCK-8 and colony formation assays were carried out to assess the biological function of PMEPA1 in PANC1 proliferation and viability. Dual-luciferase reporter assays and RT-qPCR were used to assess the interactive relationship between PMEPA1 and the MAPK signaling pathway. RESULTS By analyzing the data from GEPIA, we found that PMEPA1 mRNA expression is overexpressed in PC tissues compared with matched nontumor tissues. PMEPA1-high PC patients are predicted to have a worse prognosis than PMEPA1-low PC patients. We found that PMEPA1 shRNA suppressed PANC1 proliferation and colony formation capacity, while enforced expression of PMEPA1 yielded the opposite results. Mechanical investigations showed that PMEPA1 exerts its tumor-promoting function in pancreatic cancer via activation of the MAPK signaling pathway. CONCLUSION PMEPA1 promotes the progression of PC at least partially by activating the MAPK signaling pathway; thus, the PMEPA1/MAPK axis may be a potential therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Mengqi Song
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China; Department of Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - Bilu Li
- Department of Uroogic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - Lantian Tian
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China.
| |
Collapse
|
11
|
Bing N, Zhou H, Chen X, Hirose T, Kochi Y, Tsuchida Y, Ishigaki K, Sumitomo S, Fujio K, Zhang B, Valdez H, Vincent MS, Martin D, Clark JD. Contribution of a European-Prevalent Variant near CD83 and an East Asian-Prevalent Variant near IL17RB to Herpes Zoster Risk in Tofacitinib Treatment: Results of Genome-Wide Association Study Meta-Analyses. Arthritis Rheumatol 2021; 73:1155-1166. [PMID: 33455090 PMCID: PMC8361995 DOI: 10.1002/art.41655] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/12/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Tofacitinib is an oral JAK inhibitor for the treatment of rheumatoid arthritis (RA), psoriatic arthritis, and ulcerative colitis, and has been previously investigated for psoriasis (PsO). This meta-analysis of genome-wide association studies (GWAS) was performed to identify genetic factors associated with increased risk/faster onset of herpes zoster (HZ) in subjects with RA or PsO receiving tofacitinib treatment, and to determine potential mechanisms that could be attributed to the varying rates of HZ across ethnicities. METHODS In an ethnicity/indication-specific, trans-ethnic, trans-population meta-analysis of GWAS in subjects with RA or PsO from phase II, phase III, and long-term extension studies of tofacitinib, 8 million genetic variants were evaluated for their potential association with time to an HZ event and incidence of an HZ event (case versus control) with tofacitinib treatment, using Cox proportional hazard and logistic regression analyses, respectively. RESULTS In total, 5,246 subjects were included (3,168 with RA and 2,078 with PsO). After adjustment for age, baseline absolute lymphocyte count, genetically defined ethnicity, and concomitant methotrexate use (in RA subjects only), 4 loci were significantly associated with faster onset of HZ in European subjects (P < 5 × 10-8 ), including a single-nucleotide polymorphism (SNP) near CD83 (frequency of risk allele ~2% in European subjects versus ~0.1% in East Asian subjects). In the trans-ethnic, trans-population meta-analysis, the CD83 SNP remained significant. Four additional significant loci were identified in the meta-analysis, among which a SNP near IL17RB was associated with faster onset of HZ (meta-analysis hazard ratio 3.6 [95% confidence interval 2.40-5.44], P = 7.6 × 10-10 ; frequency of risk allele ~12% in East Asian subjects versus <0.2% in European subjects). CONCLUSION Genetic analysis of tofacitinib-treated subjects with RA or PsO identified multiple loci associated with increased HZ risk. Prevalent variants near the immune-relevant genes CD83 and IL17RB in European and East Asian populations, respectively, may contribute to risk of HZ in tofacitinib-treated subjects.
Collapse
Affiliation(s)
| | | | | | | | - Yuta Kochi
- Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan, and Medical Research Institute, Tokyo Medical and Dental UniversityTokyoJapan
| | - Yumi Tsuchida
- University of Tokyo Graduate School of MedicineTokyoJapan
| | | | - Shuji Sumitomo
- University of Tokyo Graduate School of MedicineTokyoJapan
| | - Keishi Fujio
- University of Tokyo Graduate School of MedicineTokyoJapan
| | | | | | | | | | | |
Collapse
|
12
|
Hirata H, Xu X, Nishioka K, Matsuhisa F, Kitajima S, Kukita T, Murayama M, Urano Y, Miyamoto H, Mawatari M, Kukita A. PMEPA1 and NEDD4 control the proton production of osteoclasts by regulating vesicular trafficking. FASEB J 2021; 35:e21281. [PMID: 33484199 DOI: 10.1096/fj.202001795r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/13/2020] [Accepted: 12/01/2020] [Indexed: 11/11/2022]
Abstract
Osteoclast bone resorption activity is critically regulated to maintain bone homeostasis. Osteoclasts resorb bone by producing protons and acid hydrolase via lysosomal secretion, however, a detailed mechanism remains elusive. PMEPA1 is a vesicular membrane protein, which binds to the NEDD4 family member of ubiquitin ligases. We have previously reported that Pmepa1 is highly expressed in bone resorbing osteoclasts, and regulates bone resorption. Here, we investigated the mechanism of bone resorption regulated by PMEPA1. Mutant mice lacking NEDD4-binding domains of PMEPA1 displayed enhanced bone volume, and reduced bone resorption activity in comparison with those of WT mice. Analysis with pH-sensitive fluorescence probe revealed that proton secretion from osteoclasts significantly decreased in Pmepa1 mutant osteoclasts. Immunofluorescence analysis revealed that PMEPA1 was colocalized with NEDD4, V0A3, and V0D2 subunits of vacuolar ATPase, which regulate the proton production of osteoclasts. In addition, Nedd4 knockdown reduced bone resorption and proton secretion of osteoclasts. Furthermore, Pmepa1 mutation and Nedd4 knockdown altered the cytoplasmic distribution of components of V-ATPase and expression of autophagy-related proteins, suggesting that lysosomal secretion is affected. Collectively, these findings indicate that PMEPA1 controls proton secretion from osteoclasts via NEDD4 by regulating vesicular trafficking, and NEDD4 is an important regulator of bone resorption.
Collapse
Affiliation(s)
- Hirohito Hirata
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan.,Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Xianghe Xu
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan.,Department of Molecular Cell Biology & Oral Anatomy, Kyushu University, Fukuoka, Japan
| | - Kenichi Nishioka
- Department of Internal Medicine, Musashimurayama Hospital, Tokyo, Japan
| | - Fumikazu Matsuhisa
- Division of Biological Resources and Development, Analytical Research Center for Experimental Sciences, Saga University, Saga, Japan
| | - Shuji Kitajima
- Division of Biological Resources and Development, Analytical Research Center for Experimental Sciences, Saga University, Saga, Japan
| | - Toshio Kukita
- Department of Molecular Cell Biology & Oral Anatomy, Kyushu University, Fukuoka, Japan
| | - Masatoshi Murayama
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan.,Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Yasuteru Urano
- Department of Chemical Biology & Molecular Imaging, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Chemistry & Biology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Miyamoto
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Masaaki Mawatari
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Akiko Kukita
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
13
|
Tan B, Chen Y, Xia L, Yu X, Peng Y, Zhang X, Yang Z. PMEPA1 facilitates non-small cell lung cancer progression via activating the JNK signaling pathway. Cancer Biomark 2021; 31:203-210. [PMID: 33896822 DOI: 10.3233/cbm-200966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Prostate transmembrane protein androgen-induced 1 (PMEPA1), a critical checkpoint of multiple signaling pathways, has been demonstrated to play a crucial role in various types of cancers. However, little is known about its function in non-small cell lung cancer (NSCLC). OBJECTIVE Our objective is to explore the function of PMEPA1 and its potential mechanisms in NSCLC progression. METHODS PMEPA1 expression and prognostic significance in adenocarcinoma of lung cancer (LUAD) and squamous cell carcinoma of lung cancer (LUSC) were determined using Gene Expression Profiling Interactive Analysis (GEPIA). Next, a series of cell assays were performed to examine whether overexpression or depletion of PMEPA1 affected the malignant behaviors of NSCLC H1299 cells, such as proliferation and migration. Luciferase reporter gene assays and SP600125 (a JNK inhibitor) were employed to ascertain the regulatory relationship between PMEPA1 and JNK. RESULTS PMEPA1 is overexpressed in LUAD and LUSC tissues and portends a worse prognosis for cancer patients. Gain and loss of function experiments demonstrated that PMEPA1 executes oncogenetic function in H1299 cells. Mechanism studies elucidated that PMEPA1 stimulated the transcriptional activity of the JNK pathway. CONCLUSION PMEPA1 increased the H1299 cell viability, proliferation, and migration which works, at least partially, by triggering the JNK activity. Hence, our findings support that the PMEPA1/JNK axis might be a promising therapeutic target for this challenging disease.
Collapse
|
14
|
Wang B, Zhong JL, Li HZ, Wu B, Sun DF, Jiang N, Shang J, Chen YF, Xu XH, Lu HD. Diagnostic and therapeutic values of PMEPA1 and its correlation with tumor immunity in pan-cancer. Life Sci 2021; 277:119452. [PMID: 33831430 DOI: 10.1016/j.lfs.2021.119452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/30/2022]
Abstract
AIMS The prostate transmembrane protein, androgen induced 1 (PMEPA1) is differentially expressed in pan-cancer. However, PMEPA1 specific role in cancers has not been fully clarified. This study aims to explore the potential role of Pmepa1 in pan-cancer and specific cancer, with a view to deepening the research on the pathological mechanism of cancer. MAIN METHODS The Perl language and R language were used to identify the correlation between PMEPA1 expression level and clinical indicators, prognosis values, tumor microenvironment, immune cells' infiltration, immune checkpoint genes, TMB and MSI. The Therapeutic Target Database was used for identifying potential therapeutic drugs that target the pathways that are significantly affected by PMEPA1 expression. KEY FINDINGS PMEPA1 differential expression significantly correlated with patients' age, race, tumors' stage and status. PMEPA1 high expression was closely correlated with poor prognosis in many cancer types, excluding prostate adenocarcinoma. PMEPA1 expression was closely related to tumor cells and the immune microenvironment in stromal and immune cells' level, immune cells' infiltration, immune checkpoint genes, tumor mutational burden and microsatellite instability. We also found that the activity of the olfactory transduction pathway was closely related to PMEPA1 expression. In pan-cancer, Trifluoperazine and Halofantrine have the potential to reduce PMEPA1 expression. SIGNIFICANCE This study integrated existing data to explore PMEPA1 potential function in cancers, provided insights for the future cancer-related studies.
Collapse
Affiliation(s)
- Bin Wang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Jun-Long Zhong
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Hui-Zi Li
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Biao Wu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Di-Fang Sun
- Department of Ophthalmology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
| | - Ning Jiang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Jie Shang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Yu-Feng Chen
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China
| | - Xiang-He Xu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China.
| | - Hua-Ding Lu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong, China.
| |
Collapse
|
15
|
Patel KD, Vora HH, Patel PS. Transcriptional Biomarkers in Oral Cancer: An Integrative Analysis and the Cancer Genome Atlas Validation. Asian Pac J Cancer Prev 2021; 22:371-380. [PMID: 33639650 PMCID: PMC8190349 DOI: 10.31557/apjcp.2021.22.2.371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 01/20/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE An impervious mortality rate in oral cancer (OC) to a certain extent explains the exigencies of precise biomarkers. Therefore, the study was intended to identify OC candidate biomarkers using samples of healthy normal tissues (N=335), adjacent normal tissues (N=93) and OC tissues (N=533) from online microarray data. METHODS Differentially expressed genes (DEGs) were recognised through GeneSpring software (Fold change >4.0 and 'p' value.
Collapse
Affiliation(s)
| | | | - Prabhudas S Patel
- The Gujarat Cancer & Research Institute, Civil Hospital Campus, Asarwa, Ahmedabad-380 016, Gujarat, India.
| |
Collapse
|
16
|
Molstad DHH, Zars E, Norton A, Mansky KC, Westendorf JJ, Bradley EW. Hdac3 deletion in myeloid progenitor cells enhances bone healing in females and limits osteoclast fusion via Pmepa1. Sci Rep 2020; 10:21804. [PMID: 33311522 PMCID: PMC7733476 DOI: 10.1038/s41598-020-78364-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/06/2020] [Indexed: 12/18/2022] Open
Abstract
Previous studies examining the role of the histone deacetylase Hdac3 within myeloid cells demonstrated that Hdac3 promotes M2 activation and tissue healing in inflammatory conditions. Since myeloid lineage cells are required for proper bone formation and regeneration, in this study we examined the functions of Hdac3 during bone healing. Conditional deletion of Hdac3 within myeloid progenitors accelerates healing of cortical bone defects. Moreover, reduced osteoclast numbers within the defect site are correlated with Hdac3 suppression. Ex vivo osteoclastogenesis assays further demonstrate that Hdac3 deficiency limits osteoclastogenesis, the number of nuclei per cell and bone resorption, suggesting a defect in cell fusion. High throughput RNA sequencing identified the transmembrane protein Pmepa1 as a differentially expressed gene within osteoclast progenitor cells. Knockdown of Pmepa1 partially restores defects in osteoclastogenesis induced by Hdac3 deficiency. These results show that Hdac3 is required for optimal bone healing and osteoclast fusion, potentially via its regulation of Pmepa1 expression.
Collapse
Affiliation(s)
- David H H Molstad
- Department of Orthopedics, University of Minnesota, Elizabeth W. Bradley, 100 Church St. S.E., Minneapolis, MN, 55455, USA
| | - Elizabeth Zars
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Andrew Norton
- Developmental and Surgical Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Kim C Mansky
- Developmental and Surgical Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Jennifer J Westendorf
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth W Bradley
- Department of Orthopedics, University of Minnesota, Elizabeth W. Bradley, 100 Church St. S.E., Minneapolis, MN, 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
17
|
Hagg A, Kharoud S, Goodchild G, Goodman CA, Chen JL, Thomson RE, Qian H, Gregorevic P, Harrison CA, Walton KL. TMEPAI/PMEPA1 Is a Positive Regulator of Skeletal Muscle Mass. Front Physiol 2020; 11:560225. [PMID: 33250771 PMCID: PMC7672205 DOI: 10.3389/fphys.2020.560225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/12/2020] [Indexed: 12/20/2022] Open
Abstract
Inhibition of myostatin- and activin-mediated SMAD2/3 signaling using ligand traps, such as soluble receptors, ligand-targeting propeptides and antibodies, or follistatin can increase skeletal muscle mass in healthy mice and ameliorate wasting in models of cancer cachexia and muscular dystrophy. However, clinical translation of these extracellular approaches targeting myostatin and activin has been hindered by the challenges of achieving efficacy without potential effects in other tissues. Toward the goal of developing tissue-specific myostatin/activin interventions, we explored the ability of transmembrane prostate androgen-induced (TMEPAI), an inhibitor of transforming growth factor-β (TGF-β1)-mediated SMAD2/3 signaling, to promote growth, and counter atrophy, in skeletal muscle. In this study, we show that TMEPAI can block activin A, activin B, myostatin and GDF-11 activity in vitro. To determine the physiological significance of TMEPAI, we employed Adeno-associated viral vector (AAV) delivery of a TMEPAI expression cassette to the muscles of healthy mice, which increased mass by as much as 30%, due to hypertrophy of muscle fibers. To demonstrate that TMEPAI mediates its effects via inhibition of the SMAD2/3 pathway, tibialis anterior (TA) muscles of mice were co-injected with AAV vectors expressing activin A and TMEPAI. In this setting, TMEPAI blocked skeletal muscle wasting driven by activin-induced phosphorylation of SMAD3. In a model of cancer cachexia associated with elevated circulating activin A, delivery of AAV:TMEPAI into TA muscles of mice bearing C26 colon tumors ameliorated the muscle atrophy normally associated with cancer progression. Collectively, the findings indicate that muscle-directed TMEPAI gene delivery can inactivate the activin/myostatin-SMAD3 pathway to positively regulate muscle mass in healthy settings and models of disease.
Collapse
Affiliation(s)
- Adam Hagg
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Swati Kharoud
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Georgia Goodchild
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Craig A Goodman
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science, Sunshine Hospital, The University of Melbourne, St Albans, VIC, Australia
| | - Justin L Chen
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Rachel E Thomson
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Hongwei Qian
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Paul Gregorevic
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.,Department of Neurology, The University of Washington School of Medicine, Seattle, WA, United States
| | - Craig A Harrison
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Kelly L Walton
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Hudson Institute of Medical Research, Clayton, VIC, Australia
| |
Collapse
|
18
|
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
|
19
|
Puteri MU, Watanabe Y, Wardhani BWK, Amalia R, Abdelaziz M, Kato M. PMEPA1/TMEPAI isoforms function via its PY and Smad-interaction motifs for tumorigenic activities of breast cancer cells. Genes Cells 2020; 25:375-390. [PMID: 32181976 DOI: 10.1111/gtc.12766] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/11/2020] [Accepted: 03/05/2020] [Indexed: 12/16/2022]
Abstract
PMEPA1 (prostate transmembrane protein, androgen-induced 1)/TMEPAI (transmembrane prostate androgen-induced protein) is highly expressed in diverse cancers, including breast, lung and prostate cancers. It consists of four isoforms with distinct extracellular regions (isoforms a-d). The expression and function of these isoforms are still poorly understood. Hence, we aimed to identify the preferentially expressed isoforms in breast cancer cells and analyze possible differences in tumorigenic functions. In this study, we used 5' Rapid Amplification of cDNA Ends (RACE) and Western blot analyses to identify the mRNA variants and protein isoforms of TMEPAI and found that TMEPAI isoform d as the major isoform expressed by TGF-β stimulation in breast cancer cells. We then generated CRISPR/Cas9-mediated TMEPAI knockout (KO) breast cancer cell lines and used a lentiviral expression system to complement each isoform individually. Although there were no clear functional differences between isoforms, double PPxY (PY) motifs and a Smad-interaction motif (SIM) of TMEPAI were both essential for colony and sphere formation. Collectively, our results provide a novel insight into TMEPAI isoforms in breast cancer cells and showed that coordination between double PY motifs and a SIM of TMEPAI are essential for colony and sphere formation but not for monolayer cell proliferation.
Collapse
Affiliation(s)
- Meidi U Puteri
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukihide Watanabe
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Bantari W K Wardhani
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Doctoral Program in Biomedicine, Faculty of Medicine, Universitas Indonesia, Jakarta Pusat, Indonesia
| | - Riezki Amalia
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Jawa Barat, Indonesia
| | - Mohammed Abdelaziz
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Department of Pathology, Faculty of Medicine, Sohag University, Nasr City, Egypt
| | - Mitsuyasu Kato
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
20
|
Álvarez I, Fernández I, Traoré A, Pérez-Pardal L, Menéndez-Arias NA, Goyache F. Genomic scan of selective sweeps in Djallonké (West African Dwarf) sheep shed light on adaptation to harsh environments. Sci Rep 2020; 10:2824. [PMID: 32071365 PMCID: PMC7028950 DOI: 10.1038/s41598-020-59839-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023] Open
Abstract
The Djallonké (West African Dwarf) sheep is a small-sized haired sheep resulting from a costly evolutionary process of natural adaptation to the harsh environment of West Africa including trypanosome challenge. However, genomic studies carried out in this sheep are scant. In this research, genomic data of 184 Djallonké sheep (and 12 Burkina-Sahel sheep as an outgroup) generated using medium-density SNP Chips were analyzed. Three different statistics (iHS, XP-EHH and nSL) were applied to identify candidate selection sweep regions spanning genes putatively associated with adaptation of sheep to the West African environment. A total of 207 candidate selection sweep regions were defined. Gene-annotation enrichment and functional annotation analyses allowed to identify three statistically significant functional clusters involving 12 candidate genes. Genes included in Functional Clusters associated to selection signatures were mainly related to metabolic response to stress, including regulation of oxidative and metabolic stress and thermotolerance. The bovine chromosomal areas carrying QTLs for cattle trypanotolerance were compared with the regions on which the orthologous functional candidate cattle genes were located. The importance of cattle BTA4 for trypanotolerant response might have been conserved between species. The current research provides new insights on the genomic basis for adaptation and highlights the importance of obtaining information from non-cosmopolite livestock populations managed in harsh environments.
Collapse
Affiliation(s)
- Isabel Álvarez
- Servicio Regional de Investigación y Desarrollo Agroalimentario, E-33394, Gijón, Spain
| | - Iván Fernández
- Servicio Regional de Investigación y Desarrollo Agroalimentario, E-33394, Gijón, Spain
| | - Amadou Traoré
- Institut de l'Environnement et des Recherches Agricoles (INERA), Ouagadougou, 04 BP 8645, Burkina Faso
| | | | | | - Félix Goyache
- Servicio Regional de Investigación y Desarrollo Agroalimentario, E-33394, Gijón, Spain.
| |
Collapse
|
21
|
Sharad S, Sztupinszki ZM, Chen Y, Kuo C, Ravindranath L, Szallasi Z, Petrovics G, Sreenath TL, Dobi A, Rosner IL, Srinivasan A, Srivastava S, Cullen J, Li H. Analysis of PMEPA1 Isoforms ( a and b) as Selective Inhibitors of Androgen and TGF-β Signaling Reveals Distinct Biological and Prognostic Features in Prostate Cancer. Cancers (Basel) 2019; 11:cancers11121995. [PMID: 31842254 PMCID: PMC6966662 DOI: 10.3390/cancers11121995] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 11/16/2022] Open
Abstract
Dysfunctions of androgen/TGF-β signaling play important roles in prostate tumorigenesis. Prostate Transmembrane Protein Androgen Induced 1 (PMEPA1) inhibits androgen and TGF-β signaling via a negative feedback loop. The loss of PMEPA1 confers resistance to androgen signaling inhibitors and promotes bone metastasis. Conflicting reports on the expression and biological functions of PMEPA1 in prostate and other cancers propelled us to investigate isoform specific functions in prostate cancer (PCa). One hundred and twenty laser capture micro-dissection matched normal prostate and prostate tumor tissues were analyzed for correlations between quantitative expression of PMEPA1 isoforms and clinical outcomes with Q-RT-PCR, and further validated with a The Cancer Genome Atlas (TCGA) RNA-Seq dataset of 499 PCa. Cell proliferation was assessed with cell counting, plating efficiency and soft agar assay in androgen responsive LNCaP and TGF-β responsive PC3 cells. TGF-β signaling was measured by SMAD dual-luciferase reporter assay. Higher PMEPA1-a mRNA levels indicated biochemical recurrence (p = 0.0183) and lower PMEPA1-b expression associated with metastasis (p = 0.0173). Further, lower PMEPA1-b and a higher ratio of PMEPA1-a vs. -b were correlated to higher Gleason scores and lower progression free survival rate (p < 0.01). TGF-β-responsive PMEPA1-a promoted PCa cell growth, and androgen-responsive PMEPA1-b inhibited cancer cell proliferation. PMEPA1 isoforms -a and -b were shown to be promising candidate biomarkers indicating PCa aggressiveness including earlier biochemical relapse and lower disease specific life expectancy via interrupting androgen/TGF-β signaling.
Collapse
Affiliation(s)
- Shashwat Sharad
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD 20817, USA
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Suite 100, Bethesda, MD 20817, USA
- Correspondence: (S.S.); (H.L.); Tel.: +1-240-694-4931 (S.S.); +1-240-694-4944 (H.L.)
| | | | - Yongmei Chen
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Suite 100, Bethesda, MD 20817, USA
| | - Claire Kuo
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Suite 100, Bethesda, MD 20817, USA
| | - Lakshmi Ravindranath
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Suite 100, Bethesda, MD 20817, USA
| | - Zoltan Szallasi
- Danish Cancer Society Research Center, 2100 Copenhagen, Denmark; (Z.M.S.); (Z.S.)
- Computational Health Informatics Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- SE-NAP Brain Metastasis Research group, 2nd Department of Pathology, Semmelweis University, 1085 Budapest, Hungary
| | - Gyorgy Petrovics
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD 20817, USA
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Suite 100, Bethesda, MD 20817, USA
| | - Taduru L. Sreenath
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Suite 100, Bethesda, MD 20817, USA
| | - Albert Dobi
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD 20817, USA
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Suite 100, Bethesda, MD 20817, USA
| | - Inger L. Rosner
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD 20817, USA
- Department of Urology, Walter Reed National Military Medical Center, Bethesda, MD 20814, USA
| | - Alagarsamy Srinivasan
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Suite 100, Bethesda, MD 20817, USA
| | - Shiv Srivastava
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
| | - Jennifer Cullen
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD 20817, USA
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Suite 100, Bethesda, MD 20817, USA
| | - Hua Li
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Drive, Suite 300, Bethesda, MD 20817, USA; (Y.C.); (C.K.); (L.R.); (G.P.); (T.L.S.); (A.D.); (I.L.R.); (A.S.); (J.C.)
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Suite 100, Bethesda, MD 20817, USA
- Correspondence: (S.S.); (H.L.); Tel.: +1-240-694-4931 (S.S.); +1-240-694-4944 (H.L.)
| |
Collapse
|
22
|
Lin J, Vora M, Kane NS, Gleason RJ, Padgett RW. Human Marfan and Marfan-like Syndrome associated mutations lead to altered trafficking of the Type II TGFβ receptor in Caenorhabditis elegans. PLoS One 2019; 14:e0216628. [PMID: 31071172 PMCID: PMC6508650 DOI: 10.1371/journal.pone.0216628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/24/2019] [Indexed: 12/14/2022] Open
Abstract
The transforming growth factor-β (TGFβ) family plays an important role in many developmental processes and when mutated often contributes to various diseases. Marfan syndrome is a genetic disease with an occurrence of approximately 1 in 5,000. The disease is caused by mutations in fibrillin, which lead to an increase in TGFβ ligand activity, resulting in abnormalities of connective tissues which can be life-threatening. Mutations in other components of TGFβ signaling (receptors, Smads, Schnurri) lead to similar diseases with attenuated phenotypes relative to Marfan syndrome. In particular, mutations in TGFβ receptors, most of which are clustered at the C-terminal end, result in Marfan-like (MFS-like) syndromes. Even though it was assumed that many of these receptor mutations would reduce or eliminate signaling, in many cases signaling is active. From our previous studies on receptor trafficking in C. elegans, we noticed that many of these receptor mutations that lead to Marfan-like syndromes overlap with mutations that cause mis-trafficking of the receptor, suggesting a link between Marfan-like syndromes and TGFβ receptor trafficking. To test this hypothesis, we introduced three of these key MFS and MFS-like mutations into the C. elegans TGFβ receptor and asked if receptor trafficking is altered. We find that in every case studied, mutated receptors mislocalize to the apical surface rather than basolateral surface of the polarized intestinal cells. Further, we find that these mutations result in longer animals, a phenotype due to over-stimulation of the nematode TGFβ pathway and, importantly, indicating that function of the receptor is not abrogated in these mutants. Our nematode models of Marfan syndrome suggest that MFS and MFS-like mutations in the type II receptor lead to mis-trafficking of the receptor and possibly provides an explanation for the disease, a phenomenon which might also occur in some cancers that possess the same mutations within the type II receptor (e.g. colon cancer).
Collapse
MESH Headings
- Amino Acid Sequence
- Amino Acid Substitution
- Animals
- Animals, Genetically Modified
- Caenorhabditis elegans/genetics
- Caenorhabditis elegans/metabolism
- Caenorhabditis elegans Proteins/chemistry
- Caenorhabditis elegans Proteins/genetics
- Caenorhabditis elegans Proteins/metabolism
- Disease Models, Animal
- Humans
- Marfan Syndrome/genetics
- Marfan Syndrome/metabolism
- Mutation, Missense
- Protein Domains
- Receptor, Transforming Growth Factor-beta Type II/chemistry
- Receptor, Transforming Growth Factor-beta Type II/genetics
- Receptor, Transforming Growth Factor-beta Type II/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Transforming Growth Factor beta/chemistry
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Sequence Homology, Amino Acid
- Species Specificity
Collapse
Affiliation(s)
- Jing Lin
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
| | - Mehul Vora
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
- * E-mail: (MV); (RWP)
| | - Nanci S. Kane
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
| | - Ryan J. Gleason
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Richard W. Padgett
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
- * E-mail: (MV); (RWP)
| |
Collapse
|
23
|
H3.3 Barcoding of Nucleus Accumbens Transcriptional Activity Identifies Novel Molecular Cascades Associated with Cocaine Self-administration in Mice. J Neurosci 2019; 39:5247-5254. [PMID: 31043484 DOI: 10.1523/jneurosci.0015-19.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/17/2019] [Accepted: 04/26/2019] [Indexed: 02/06/2023] Open
Abstract
Although numerous epigenetic modifications have been associated with addiction, little work has explored the turnover of histone variants. Uniquely, the H3.3 variant incorporates stably and preferentially into chromatin independently of DNA replication at active sites of transcription and transcription factor binding. Thus, genomic regions associated with H3.3-containing nucleosomes are particularly likely to be involved in plasticity, such as following repeated cocaine exposure. A recently developed mouse line expressing a neuron-specific hemagglutinin (HA)-tagged H3.3 protein was used to track transcriptionally active sites cumulatively across 19 d of cocaine self-administration. RNA-seq and H3.3-HA ChIP-seq analyses were performed on NAcc tissue collected following cocaine or food self-administration in male mice. RNA sequencing revealed five genes upregulated in cocaine relative to food self-administering mice: Fosb, Npas4, Vgf, Nptx2, and Pmepa1, which reflect known and novel cocaine plasticity-associated genes. Subsequent ChIP-seq analysis confirmed increased H3.3 aggregation at four of these five loci, thus validating H3.3 insertion as a marker of enhanced cocaine-induced transcription. Further motif recognition analysis of the ChIP-seq data showed that cocaine-associated differential H3.3 accumulation correlated with the presence of several transcription factor binding motifs, including RBPJ1, EGR1, and SOX4, suggesting that these are potentially important regulators of molecular cascades associated with cocaine-induced neuronal plasticity. Additional ontological analysis revealed differential H3.3 accumulation mainly near genes involved in neuronal differentiation and dendrite formation. These results establish the H3.3-HA transgenic mouse line as a compelling molecular barcoding tool to identify the cumulative effects of long-term environmental perturbations, such as exposure to drugs of abuse.SIGNIFICANCE STATEMENT Histone H3.3 is a core histone variant that is stably incorporated at active sites of transcription. We used a tagged version of H3.3 expressed exclusively in neurons to delineate active transcription sites following extended cocaine self-administration in mice. This approach revealed the cumulative list of genes expressed in response to cocaine taking over the course of several weeks. We combined this technique with RNA sequencing of tissue collected from the same animals 24 h after the last cocaine exposure. Comparing these datasets provided a full picture of genes that respond to chronic cocaine exposure in NAcc neurons. These studies revealed novel transcription factors that are likely involved in cocaine-induced plasticity and addiction-like behaviors.
Collapse
|
24
|
Ikeno S, Nakano N, Sano K, Minowa T, Sato W, Akatsu R, Sakata N, Hanagata N, Fujii M, Itoh F, Itoh S. PDZK1-interacting protein 1 (PDZK1IP1) traps Smad4 protein and suppresses transforming growth factor-β (TGF-β) signaling. J Biol Chem 2019; 294:4966-4980. [PMID: 30718277 DOI: 10.1074/jbc.ra118.004153] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 12/27/2018] [Indexed: 01/10/2023] Open
Abstract
Transforming growth factor (TGF)-β signaling in humans is stringently regulated to prevent excessive TGF-β signaling. In tumors, TGF-β signaling can both negatively and positively regulate tumorigenesis dependent on tumor type, but the reason for these opposite effects is unclear. TGF-β signaling is mainly mediated via the Smad-dependent pathway, and herein we found that PDZK1-interacting protein 1 (PDZK1IP1) interacts with Smad4. PDZK1IP1 inhibited both the TGF-β and the bone morphogenetic protein (BMP) pathways without affecting receptor-regulated Smad (R-Smad) phosphorylation. Rather than targeting R-Smad phosphorylation, PDZK1IP1 could interfere with TGF-β- and BMP-induced R-Smad/Smad4 complex formation. Of note, PDZK1IP1 retained Smad4 in the cytoplasm of TGF-β-stimulated cells. To pinpoint PDZK1IP1's functional domain, we created several PDZK1IP1 variants and found that its middle region, from Phe40 to Ala49, plays a key role in its Smad4-regulating activity. PDZK1IP1 knockdown enhanced the expression of the TGF-β target genes Smad7 and prostate transmembrane protein androgen-induced (TMEPAI) upon TGF-β stimulation. In contrast, PDZK1IP1 overexpression suppressed TGF-β-induced reporter activities, cell migration, and cell growth inhibition. In a xenograft tumor model in which TGF-β was previously shown to elicit tumor-promoting effects, PDZK1IP1 gain of function decreased tumor size and increased survival rates. Taken together, these findings indicate that PDZK1IP1 interacts with Smad4 and thereby suppresses the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Souichi Ikeno
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Naoko Nakano
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Keigo Sano
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Takashi Minowa
- Nanotechnology Innovation Station, National Institute of Materials Science, Tsukuba, Ibaraki 305-0047, Japan
| | - Wataru Sato
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Ryosuke Akatsu
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Nobuo Sakata
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Nobutaka Hanagata
- Nanotechnology Innovation Station, National Institute of Materials Science, Tsukuba, Ibaraki 305-0047, Japan
| | - Makiko Fujii
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan, and
| | - Fumiko Itoh
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Susumu Itoh
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan,
| |
Collapse
|