1
|
Schibalski RS, Shulha AS, Tsao BP, Palygin O, Ilatovskaya DV. The role of polyamine metabolism in cellular function and physiology. Am J Physiol Cell Physiol 2024; 327:C341-C356. [PMID: 38881422 PMCID: PMC11427016 DOI: 10.1152/ajpcell.00074.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Polyamines are molecules with multiple amino groups that are essential for cellular function. The major polyamines are putrescine, spermidine, spermine, and cadaverine. Polyamines are important for posttranscriptional regulation, autophagy, programmed cell death, proliferation, redox homeostasis, and ion channel function. Their levels are tightly controlled. High levels of polyamines are associated with proliferative pathologies such as cancer, whereas low polyamine levels are observed in aging, and elevated polyamine turnover enhances oxidative stress. Polyamine metabolism is implicated in several pathophysiological processes in the nervous, immune, and cardiovascular systems. Currently, manipulating polyamine levels is under investigation as a potential preventive treatment for several pathologies, including aging, ischemia/reperfusion injury, pulmonary hypertension, and cancer. Although polyamines have been implicated in many intracellular mechanisms, our understanding of these processes remains incomplete and is a topic of ongoing investigation. Here, we discuss the regulation and cellular functions of polyamines, their role in physiology and pathology, and emphasize the current gaps in knowledge and potential future research directions.
Collapse
Affiliation(s)
- Ryan S Schibalski
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Anastasia S Shulha
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Betty P Tsao
- Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
2
|
Zhou M, Yin X, Chen B, Hu S, Zhou W. A PET probe targeting polyamine transport system for precise tumor diagnosis and therapy. Asian J Pharm Sci 2024; 19:100924. [PMID: 38903130 PMCID: PMC11186966 DOI: 10.1016/j.ajps.2024.100924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/02/2024] [Accepted: 03/04/2024] [Indexed: 06/22/2024] Open
Abstract
Polyamine metabolism dysregulation is a hallmark of many cancers, offering a promising avenue for early tumor theranostics. This study presents the development of a nuclear probe derived from spermidine (SPM) for dual-purpose tumor PET imaging and internal radiation therapy. The probe, radiolabeled with either [68Ga]Ga for diagnostic applications or [177Lu]Lu for therapeutic use, was synthesized with exceptional purity, stability, and specific activity. Extensive testing involving 12 different tumor cell lines revealed remarkable specificity towards B16 melanoma cells, showcasing outstanding tumor localization and target-to-non-target ratio. Mechanistic investigations employing polyamines, non-labeled precursor, and polyamine transport system (PTS) inhibitor, consistently affirmed the probe's targetability through recognition of the PTS. Notably, while previous reports indicated PTS upregulation in various tumor types for targeted therapy, this study observed no positive signals, highlighting a concentration-dependent discrepancy between targeting for therapy and diagnosis. Furthermore, when labeled with [177Lu], the probe demonstrated its therapeutic potential by effectively controlling tumor growth and extending mouse survival. Investigations into biodistribution, excretion, and biosafety in healthy humans laid a robust foundation for clinical translation. This study introduces a versatile SPM-based nuclear probe with applications in precise tumor theranostics, offering promising prospects for clinical implementation.
Collapse
Affiliation(s)
- Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Xiaoqin Yin
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Bei Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha 410008, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha 410008, China
| |
Collapse
|
3
|
Du M, Meng X, Zhou B, Song W, Shi J, Liang M, Liang Y, Gao Y. A risk score based on polyamine metabolism and chemotherapy-related genes predicts prognosis and immune cells infiltration of lung adenocarcinoma. J Cell Mol Med 2024; 28:e18387. [PMID: 38924680 PMCID: PMC11200130 DOI: 10.1111/jcmm.18387] [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: 12/06/2023] [Revised: 04/16/2024] [Accepted: 04/27/2024] [Indexed: 06/28/2024] Open
Abstract
We aimed to explore whether the genes associated with both platinum-based therapy and polyamine metabolism could predict the prognosis of LUAD. We searched for the differential expression genes (DEGs) associated with platinum-based therapy, then we interacted them with polyamine metabolism-related genes to obtain hub genes. Subsequently, we analysed the main immune cell populations in LUAD using the scRNA-seq data, and evaluated the activity of polyamine metabolism of different cell subpopulations. The DEGs between high and low activity groups were screened to identify key DEGs to establish prognostic risk score model. We further elucidated the landscape of immune cells, mutation and drug sensitivity analysis in different risk groups. Finally, we got 10 hub genes associated with both platinum-based chemotherapy and polyamine metabolism, and found that these hub genes mainly affected signalling transduction pathways. B cells and mast cells with highest polyamine metabolism activity, while NK cells were found with lowest polyamine metabolism activity based on scRNA-seq data. DEGs between high and low polyamine metabolism activity groups were identified, then 6 key genes were screened out to build risk score, which showed a good predictive power. The risk score showed a universal negative correlation with immunotherapy checkpoint genes and the cytotoxic T cells infiltration. The mutation rates of EGFR in low-risk group was significantly higher than that of high-risk group. In conclusion, we developed a risk score based on key genes associated with platinum-based therapy and polyamine metabolism, which provide a new perspective for prognosis prediction of LUAD.
Collapse
Affiliation(s)
- Minjun Du
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiangzhi Meng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Boxuan Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Weijian Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jianwei Shi
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Mei Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yicheng Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of thoracic surgery, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
4
|
Tan B, Xiao D, Wang J, Tan B. The Roles of Polyamines in Intestinal Development and Function in Piglets. Animals (Basel) 2024; 14:1228. [PMID: 38672376 PMCID: PMC11047586 DOI: 10.3390/ani14081228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
The gastrointestinal tract plays crucial roles in the digestion and absorption of nutrients, as well as in maintenance of a functional barrier. The development and maturation of the intestine is important for piglets to maintain optimal growth and health. Polyamines are necessary for the proliferation and growth of enterocytes, which play a key role in differentiation, migration, remodeling and integrity of the intestinal mucosa after injury. This review elaborates the development of the structure and function of the intestine of piglets during embryonic, suckling and weaning periods, the utilization and metabolism of polyamines in the intestine, as well as the role of polyamines in intestinal development and mucosal repair. The nutritional intervention to improve intestinal development and functions by modulating polyamine metabolism in piglets is also put forward. These results may help to promote the adaption to weaning in pigs and provide useful information for the development and health of piglets.
Collapse
Affiliation(s)
- Bihui Tan
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (B.T.); (D.X.); (J.W.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Dingfu Xiao
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (B.T.); (D.X.); (J.W.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Jing Wang
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (B.T.); (D.X.); (J.W.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Bi’e Tan
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (B.T.); (D.X.); (J.W.)
- Yuelushan Laboratory, Changsha 410128, China
- Hunan Linxi Biological Technology Co., Ltd. Expert Workstation, Changsha 410202, China
| |
Collapse
|
5
|
Sadat Kalaki N, Ahmadzadeh M, Najafi M, Mobasheri M, Ajdarkosh H, Karbalaie Niya MH. Systems biology approach to identify biomarkers and therapeutic targets for colorectal cancer. Biochem Biophys Rep 2024; 37:101633. [PMID: 38283191 PMCID: PMC10821538 DOI: 10.1016/j.bbrep.2023.101633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 01/30/2024] Open
Abstract
Background Colorectal cancer (CRC), is the third most prevalent cancer across the globe, and is often detected at advanced stage. Late diagnosis of CRC, leave the chemotherapy and radiotherapy as the main options for the possible treatment of the disease which are associated with severe side effects. In the present study, we seek to explore CRC gene expression data using a systems biology framework to identify potential biomarkers and therapeutic targets for earlier diagnosis and treatment of the disease. Methods The expression data was retrieved from the gene expression omnibus (GEO). Differential gene expression analysis was conducted using R/Bioconductor package. The PPI network was reconstructed by the STRING. Cystoscope and Gephi software packages were used for visualization and centrality analysis of the PPI network. Clustering analysis of the PPI network was carried out using k-mean algorithm. Gene-set enrichment based on Gene Ontology (GO) and KEGG pathway databases was carried out to identify the biological functions and pathways associated with gene groups. Prognostic value of the selected identified hub genes was examined by survival analysis, using GEPIA. Results A total of 848 differentially expressed genes were identified. Centrality analysis of the PPI network resulted in identification of 99 hubs genes. Clustering analysis dissected the PPI network into seven interactive modules. While several DEGs and the central genes in each module have already reported to contribute to CRC progression, survival analysis confirmed high expression of central genes, CCNA2, CD44, and ACAN contribute to poor prognosis of CRC patients. In addition, high expression of TUBA8, AMPD3, TRPC1, ARHGAP6, JPH3, DYRK1A and ACTA1 was found to associate with decreased survival rate. Conclusion Our results identified several genes with high centrality in PPI network that contribute to progression of CRC. The fact that several of the identified genes have already been reported to be relevant to diagnosis and treatment of CRC, other highlighted genes with limited literature information may hold potential to be explored in the context of CRC biomarker and drug target discovery.
Collapse
Affiliation(s)
- Niloufar Sadat Kalaki
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
- International Institute of New Sciences (IINS), Tehran, Iran
| | - Mozhgan Ahmadzadeh
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohammad Najafi
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Meysam Mobasheri
- Department of Biotechnology, Faculty of Advanced Sciences and Technology, Tehran Islamic Azad University of Medical Sciences, Tehran, Iran
- International Institute of New Sciences (IINS), Tehran, Iran
| | - Hossein Ajdarkosh
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hadi Karbalaie Niya
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Pérez-Riesgo E, Hernando-Pérez E, Feijóo V, Tajada S, Núñez L, Villalobos C. Transcriptional Basis of Ca 2+ Remodeling Reversal Induced by Polyamine Synthesis Inhibition in Colorectal Cancer Cells. Cancers (Basel) 2023; 15:cancers15051600. [PMID: 36900391 PMCID: PMC10000432 DOI: 10.3390/cancers15051600] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Colorectal cancer (CRC) is associated with mutations in APC/Wnt leading to c-myc activation and the overexpression of ODC1, the limiting step in polyamine synthesis. CRC cells also display a remodeling of intracellular Ca2+ homeostasis that contributes to cancer hallmarks. As polyamines may modulate Ca2+ homeostasis during epithelial tissue repair, we investigated whether polyamine synthesis inhibition may reverse Ca2+ remodeling in CRC cells and, if so, the molecular basis for this reversal. To this end, we used calcium imaging and transcriptomic analysis in normal and CRC cells treated with DFMO, an ODC1 suicide inhibitor. We found that polyamine synthesis inhibition partially reversed changes in Ca2+ homeostasis associated with CRC, including a decrease in resting Ca2+ and SOCE along with an increased Ca2+ store content. We also found that polyamine synthesis inhibition reversed transcriptomic changes in CRC cells without affecting normal cells. Specifically, DFMO treatment enhanced the transcription of SOCE modulators CRACR2A; ORMDL3; and SEPTINS 6, 7, 8, 9, and 11, whereas it decreased SPCA2, involved in store-independent Orai1 activation. Therefore, DFMO treatment probably decreased store-independent Ca2+ entry and enhanced SOCE control. Conversely, DFMO treatment decreased the transcription of the TRP channels TRPC1 and 5, TRPV6, and TRPP1 while increasing TRPP2, thus probably decreasing Ca2+ entry through TRP channels. Finally, DFMO treatment enhanced the transcription of the PMCA4 Ca2+ pump and mitochondrial channels MCU and VDAC3 for enhanced Ca2+ extrusion through the plasma membrane and mitochondria. Collectively, these findings suggested the critical role of polyamines in Ca2+ remodeling in colorectal cancer.
Collapse
Affiliation(s)
- Enrique Pérez-Riesgo
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain
- Correspondence: (E.P.-R.); (C.V.); Tel.: +34-983-184822 (E.P.-R.); +34-983-184821 (C.V.)
| | - Elena Hernando-Pérez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain
| | - Verónica Feijóo
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain
| | - Sendoa Tajada
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Lucía Núñez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Carlos Villalobos
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain
- Correspondence: (E.P.-R.); (C.V.); Tel.: +34-983-184822 (E.P.-R.); +34-983-184821 (C.V.)
| |
Collapse
|
7
|
Mo W, Liu G, Wu C, Jia G, Zhao H, Chen X, Wang J. STIM1 promotes IPEC-J2 porcine epithelial cell restitution by TRPC1 signaling. Anim Biotechnol 2022; 33:1492-1503. [PMID: 33866928 DOI: 10.1080/10495398.2021.1910044] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Intestinal epithelial restitution is partly dependent on cell migration, which reseals superficial wounding after injury. Here, we tested the hypothesis that stromal interaction molecule 1(STIM1) regulates porcine intestinal epithelial cell migration by activating transient receptor potential canonical 1 (TRPC1) signaling. Results showed that the knockdown of STIM1 repressed cell migration after wounding, reduced the protein concentration of STIM1 and TRPC1, and decreased the inositol trisphosphate (IP3) content in IPEC-J2 cells (p < 0.05). However, overexpression of STIM1 obtained opposite results (p < 0.05). The inhibition of TRPC1 activity by treatment with SKF96365 in cells overexpressing wild-type and mutant STIM1 attenuated the STIM1 overexpression-induced increase of cell migration, STIM1, TRPC1 and IP3 (p < 0.05). In addition, polyamine depletion caused by α-difluoromethylornithine (DFMO) resulted in the decrease of above-mentioned parameters, and exogenous polyamine could attenuate the negative effects of DFMO on IPEC-J2 cells (p < 0.05). Moreover, the overexpression of STIM1 could rescue cell migration, the protein level of STIM1 and TRPC1, and IP3 content in polyamine-deficient IPEC-J2 cells (p < 0.05). These results indicated that STIM1 could enhance porcine intestinal epithelial cell migration via the TRPC1 signaling pathway. Inhibition of cell migration by polyamine depletion resulted from the reduction of STIM1 activity.
Collapse
Affiliation(s)
- Weiwei Mo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Guangmang Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Caimei Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Gang Jia
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Hua Zhao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Jing Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
8
|
An Update on the Metabolic Landscape of Oncogenic Viruses. Cancers (Basel) 2022; 14:cancers14235742. [PMID: 36497226 PMCID: PMC9738352 DOI: 10.3390/cancers14235742] [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: 10/10/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Viruses play an important role in cancer development as about 12% of cancer types are linked to viral infections. Viruses that induce cellular transformation are known as oncoviruses. Although the mechanisms of viral oncogenesis differ between viruses, all oncogenic viruses share the ability to establish persistent chronic infections with no obvious symptoms for years. During these prolonged infections, oncogenic viruses manipulate cell signaling pathways that control cell cycle progression, apoptosis, inflammation, and metabolism. Importantly, it seems that most oncoviruses depend on these changes for their persistence and amplification. Metabolic changes induced by oncoviruses share many common features with cancer metabolism. Indeed, viruses, like proliferating cancer cells, require increased biosynthetic precursors for virion production, need to balance cellular redox homeostasis, and need to ensure host cell survival in a given tissue microenvironment. Thus, like for cancer cells, viral replication and persistence of infected cells frequently depend on metabolic changes. Here, we draw parallels between metabolic changes observed in cancers or induced by oncoviruses, with a focus on pathways involved in the regulation of glucose, lipid, and amino acids. We describe whether and how oncoviruses depend on metabolic changes, with the perspective of targeting them for antiviral and onco-therapeutic approaches in the context of viral infections.
Collapse
|
9
|
Lian J, Liang Y, Zhang H, Lan M, Ye Z, Lin B, Qiu X, Zeng J. The role of polyamine metabolism in remodeling immune responses and blocking therapy within the tumor immune microenvironment. Front Immunol 2022; 13:912279. [PMID: 36119047 PMCID: PMC9479087 DOI: 10.3389/fimmu.2022.912279] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
The study of metabolism provides important information for understanding the biological basis of cancer cells and the defects of cancer treatment. Disorders of polyamine metabolism is a common metabolic change in cancer. With the deepening of understanding of polyamine metabolism, including molecular functions and changes in cancer, polyamine metabolism as a new anti-cancer strategy has become the focus of attention. There are many kinds of polyamine biosynthesis inhibitors and transport inhibitors, but not many drugs have been put into clinical application. Recent evidence shows that polyamine metabolism plays essential roles in remodeling the tumor immune microenvironment (TIME), particularly treatment of DFMO, an inhibitor of ODC, alters the immune cell population in the tumor microenvironment. Tumor immunosuppression is a major problem in cancer treatment. More and more studies have shown that the immunosuppressive effect of polyamines can help cancer cells to evade immune surveillance and promote tumor development and progression. Therefore, targeting polyamine metabolic pathways is expected to become a new avenue for immunotherapy for cancer.
Collapse
Affiliation(s)
- Jiachun Lian
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yanfang Liang
- Department of Pathology, Dongguan Hospital Affiliated to Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Hailiang Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Minsheng Lan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Ziyu Ye
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Department of Pathology, Dongguan Hospital Affiliated to Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- Dongguan Metabolite Analysis Engineering Technology Center of Cells for Medical Use, Guangdong Xinghai Institute of Cell, Dongguan, China
| | - Bihua Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xianxiu Qiu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| | - Jincheng Zeng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Dongguan Metabolite Analysis Engineering Technology Center of Cells for Medical Use, Guangdong Xinghai Institute of Cell, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
10
|
Aspalathus linearis (Rooibos) and Agmatine May Act Synergistically to Beneficially Modulate Intestinal Tight Junction Integrity and Inflammatory Profile. Pharmaceuticals (Basel) 2022; 15:ph15091097. [PMID: 36145318 PMCID: PMC9501288 DOI: 10.3390/ph15091097] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/17/2022] [Accepted: 08/28/2022] [Indexed: 11/25/2022] Open
Abstract
In order to promote gastrointestinal health, significant increases in the prevalence of gastrointestinal disorders should be paralleled by similar surges in therapeutics research. Nutraceutical interventions may play a significant role in patient management. The current study aimed to determine the potential of Aspalathus linearis (rooibos) to prevent gastrointestinal dysregulation resulting from high-dose trace-amine (TA) exposure. Considering the substantial female bias in functional gastrointestinal disorders, and the suggested phytoestrogenicity of rooibos, the study design allowed for a comparison between the effects of an ethanol extract of green rooibos and 17β-estradiol (E2). High levels of ρ-tyramine (TYR) and agmatine (AGM), but not β-phenethylamine (PEA) or tryptamine (TRP), resulted in prostaglandin E2 (PGE2) hypersecretion, increased tight-junction protein (TJP; occludin and ZO-1) secretion and (dissimilarly) disrupted the TJP cellular distribution profile. Modulating benefits of rooibos and E2 were TA-specific. Rooibos pre-treatment generally reduced IL-8 secretion across all TA conditions and prevented PGE2 hypersecretion after exposure to both TYR and AGM, but was only able to normalise TJP levels and the distribution profile in AGM-exposed cells. In contrast, E2 pre-treatment prevented only TYR-associated PGE2 hypersecretion and TJP dysregulation. Together, the data suggest that the antioxidant and anti-inflammatory effects of rooibos, rather than phytoestrogenicity, affect benefits illustrated for rooibos.
Collapse
|
11
|
Bravo Iniguez A, Tian Q, Du M, Zhu MJ. Alpha-Ketoglutarate Promotes Goblet Cell Differentiation and Alters Urea Cycle Metabolites in DSS-Induced Colitis Mice. Nutrients 2022; 14:nu14061148. [PMID: 35334805 PMCID: PMC8951758 DOI: 10.3390/nu14061148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/23/2022] [Accepted: 03/04/2022] [Indexed: 12/04/2022] Open
Abstract
The metabolite, alpha-ketoglutarate (aKG), shows promise as an approach for ameliorating colitis, but much remains unknown about the full extent of its effects on the metabolome and mucosal barrier. To further elucidate this matter, C57BL/6 male mice received drinking water with or without 1% aKG for three weeks, then were subjected to 2.5% dextran sulfate sodium (DSS) induction for 7 days followed by 7 days of recovery. Cecal content and intestinal tissue samples were analyzed for changes in metabolite profile and signaling pathways. Gas chromatography-mass spectrometry (GC-MS) metabolomics revealed a separation between the metabolome of mice treated with or without aKG; putrescine and glycine were significantly increased; and ornithine and amide products, oleamide and urea were significantly decreased. Based on a pathway analysis, aKG treatment induced metabolite changes and enriched glutathione metabolism and the urea cycle. Additionally, signaling pathways committing epithelial cells to the secretory lineage were elevated in aKG-treated mice. Consistently, aKG supplementation increased goblet cells staining, mRNA expression of mucin 2, and, trefoil factor 3 and Krüppel-like factor 4, markers of goblet cell differentiation. These data suggest the ameliorating the effects of aKG against chemically induced colitis involves a reduction in harmful metabolites and the promotion of goblet cell differentiation, resulting in a more-fortified mucus layer.
Collapse
Affiliation(s)
- Alejandro Bravo Iniguez
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.T.)
| | - Qiyu Tian
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.T.)
- Department of Animal Science, Washington State University, Pullman, WA 99164, USA;
| | - Min Du
- Department of Animal Science, Washington State University, Pullman, WA 99164, USA;
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.T.)
- Correspondence: ; Tel.: +1-509-335-4016
| |
Collapse
|
12
|
Wang SR, Rathor N, Kwon MS, Xiao L, Chung HK, Turner DJ, Wang JY, Rao JN. miR-195 Regulates Intestinal Epithelial Restitution after Wounding by altering Actin-Related Protein-2 Translation. Am J Physiol Cell Physiol 2022; 322:C712-C722. [PMID: 35235424 PMCID: PMC8977142 DOI: 10.1152/ajpcell.00001.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Early gut epithelial restitution reseals superficial wounds after acute injury, but the exact mechanism underlying this rapid mucosal repair remains largely unknown. MicroRNA-195 (miR-195) is highly expressed in the gut epithelium and involved in many aspects of mucosal pathobiology. Actin-related proteins (ARPs) are key components essential for stimulation of actin polymerization and regulate cell motility. Here we reported that miR-195 modulates early intestinal epithelial restitution by altering ARP-2 expression at the translation level. MiR-195 directly interacted with the ARP-2 mRNA, and ectopically overexpressed miR-195 decreased ARP-2 protein without effect on its mRNA content. In contrast, miR-195 silencing by transfection with the anti-miR-195 increased ARP-2 protein expression. Decreased ARP-2 levels by miR-195 were associated with an inhibition of early epithelial restitution, as indicated by a decrease in cell migration over the wounded area. Elevation of cellular ARP-2 levels by transfection with its transgene restored cell migration after wounding in cells overexpressing miR-195. Polyamines were found to decrease miR-195 abundance and enhanced ARP-2 translation, thus promoting epithelial restitution after wounding. Moreover, increasing the levels of miR-195 disrupted F-actin cytoskeleton organization, which was prevented by ARP2 overexpression. These results indicate that miR-195 inhibits early epithelial restitution by decreasing ARP-2 translation and that miR-195 expression is negatively regulated by cellular polyamines.
Collapse
Affiliation(s)
- Shelley R Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Cell Biology Group, Baltimore VA Medical Center, Baltimore, MD, United States
| | - Navneeta Rathor
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Min S Kwon
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Cell Biology Group, Baltimore VA Medical Center, Baltimore, MD, United States
| | - Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Cell Biology Group, Baltimore VA Medical Center, Baltimore, MD, United States
| | - Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Cell Biology Group, Baltimore VA Medical Center, Baltimore, MD, United States
| | - Douglas J Turner
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Cell Biology Group, Baltimore VA Medical Center, Baltimore, MD, United States
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Cell Biology Group, Baltimore VA Medical Center, Baltimore, MD, United States.,Cell Biology Group, Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Cell Biology Group, Baltimore VA Medical Center, Baltimore, MD, United States
| |
Collapse
|
13
|
Xiao W, Zhang Q, Yu L, Tian F, Chen W, Zhai Q. Effects of vegetarian diet-associated nutrients on gut microbiota and intestinal physiology. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2021.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
14
|
Pretorius L, Van Staden ADP, Van der Merwe JJ, Henning N, Smith C. Alterations to microbial secretome by estrogen may contribute to sex bias in irritable bowel syndrome. Inflammopharmacology 2022; 30:267-281. [PMID: 35022916 DOI: 10.1007/s10787-021-00906-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Irritable bowel syndrome (IBS) is a female predominant functional gastrointestinal disorder, underpinned by microbial dysbiosis and microinflammation. We suggest that changes in trace amine (TA) load and metabolism may link together diet, inflammation and sex in this context. METHODS The effect of E2 treatment on microbial growth and TA generation was assessed using liquid chromatography and tandem mass spectrometry methodology. To investigate the effects of TAs on the gut, WST-1, prostaglandin E2 and tight junction protein dynamics were investigated in TA treated (HT-29) colon epithelial monolayer cultures. RESULTS Differential E2-dependent alterations of the TA production capabilities of microbes were observed. Significantly, E2 treatment resulted in a 50% increase in tryptamine secretion from a probiotic microbe (p < 0.0001). Moreover, in vitro experiments indicated that TA treatment exerted type-specific effects in the gut, e.g., reducing mitochondrial functionality, even at low doses of tryptamine (p < 0.0001) and ρ-tyramine (p < 0.001). Additionally, prostaglandin E2 levels were significantly increased following ρ-tyramine and agmatine treatment (p < 0.05). In terms of functionality, all investigated TAs resulted in occludin redistribution and loss of zona occludens-1 and occludin co-localization. CONCLUSION Increases in the gastrointestinal TA load may contribute to a relatively pro-inflammatory outcome in the intestine, along with tight junction protein disruption. Additionally, fluctuating levels of endogenous E2 may modulate microbially-derived TA levels, potentially explaining exaggerating gastrointestinal symptomology in females during low E2 phases. Thus, current data warrants subsequent investigations in appropriate in vivo models to fully elucidate the role of the trace aminergic system in the sex bias observed in IBS.
Collapse
Affiliation(s)
- Lesha Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Anton du Preez Van Staden
- Department of Microbiology, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Johannes J Van der Merwe
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa.,LabSPACE, Midrand, South Africa
| | - Natasha Henning
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Carine Smith
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
15
|
Shalygin A, Kolesnikov D, Glushankova L, Gusev K, Skopin A, Skobeleva K, Kaznacheyeva EV. Role of STIM2 and Orai proteins in regulating TRPC1 channel activity upon calcium store depletion. Cell Calcium 2021; 97:102432. [PMID: 34157631 DOI: 10.1016/j.ceca.2021.102432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 05/08/2021] [Accepted: 06/04/2021] [Indexed: 11/17/2022]
Abstract
Store-operated calcium channels are the major player in calcium signaling in non-excitable cells. Store-operated calcium entry is associated with the Orai, stromal interaction molecule (STIM), and transient receptor potential canonical (TRPC) protein families. Researchers have provided conflicting data about TRPC1 channel regulation by Orai and STIM. To determine how Orai and STIM influence endogenous TRPC1 pore properties and regulation, we used single channel patch-clamp recordings. Here we showed that knockout or knockdown of Orai1 or Orai3 or overexpression of the dominant-negative mutant Orai1 E106Q did not change the conductance or selectivity of single TRPC1 channels. In addition, these TRPC1 channel properties did not depend on the amount of STIM1 and STIM2 proteins. To study STIM2-mediated regulation of TRPC1 channels, we utilized partial calcium store depletion induced by application of 10 nM thapsigargin (Tg). TRPC1 activation by endogenous STIM2 was greatly decreased in acute extracellular calcium-free experiments. STIM2 overexpression increased both the basal activity and number of silent TRPC1 channels in the plasma membrane. After calcium store depletion, overexpressed STIM2 directly activated TRPC1 in the plasma membrane even without calcium entry in acute experiments. However, this effect was abrogated by co-expression with the non-permeable Orai1 E106Q mutant protein. Taken together, our single-channel patch clamp experiments clearly demonstrated that endogenous TRPC1 forms a channel pore without involving Orai proteins. Calcium entry through Orai triggered TRPC1 channel activation in the plasma membrane, while subsequent STIM2-mediated TRPC1 activity regulation was not dependent on calcium entry.
Collapse
Affiliation(s)
- A Shalygin
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia.
| | - D Kolesnikov
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - L Glushankova
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - K Gusev
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - A Skopin
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - K Skobeleva
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - E V Kaznacheyeva
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia.
| |
Collapse
|
16
|
Chandwe K, Kelly P. Colostrum Therapy for Human Gastrointestinal Health and Disease. Nutrients 2021; 13:1956. [PMID: 34200282 PMCID: PMC8228205 DOI: 10.3390/nu13061956] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/12/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
There is increasing awareness that a broad range of gastrointestinal diseases, and some systemic diseases, are characterized by failure of the mucosal barrier. Bovine colostrum is a complex biological fluid replete with growth factors, nutrients, hormones, and paracrine factors which have a range of properties likely to contribute to mucosal healing in a wide range of infective, inflammatory, and injury conditions. In this review, we describe the anatomy and physiology of the intestinal barrier and how it may fail. We survey selected diseases in which disordered barrier function contributes to disease pathogenesis or progression, and review the evidence for or against efficacy of bovine colostrum in management. These disorders include enteropathy due to non-steroidal anti-inflammatory drugs (NSAIDs), inflammatory bowel disease (IBD), necrotizing enterocolitis, infectious diarrhea, intestinal failure, and damage due to cancer therapy. In animal models, bovine colostrum benefits NSAID enteropathy, IBD, and intestinal failure. In human trials, there is substantial evidence of efficacy of bovine colostrum in inflammatory bowel disease and in infectious diarrhea. Given the robust scientific rationale for using bovine colostrum as a promoter of mucosal healing, further work is needed to define its role in therapy.
Collapse
Affiliation(s)
- Kanta Chandwe
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka 10101, Zambia;
| | - Paul Kelly
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka 10101, Zambia;
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
17
|
Rathor N, Chung HK, Song JL, Wang SR, Wang JY, Rao JN. TRPC1-mediated Ca 2+ signaling enhances intestinal epithelial restitution by increasing α4 association with PP2Ac after wounding. Physiol Rep 2021; 9:e14864. [PMID: 33991460 PMCID: PMC8123541 DOI: 10.14814/phy2.14864] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/02/2021] [Accepted: 01/13/2021] [Indexed: 11/24/2022] Open
Abstract
Gut epithelial restitution after superficial wounding is an important repair modality regulated by numerous factors including Ca2+ signaling and cellular polyamines. Transient receptor potential canonical-1 (TRPC1) functions as a store-operated Ca2+ channel in intestinal epithelial cells (IECs) and its activation increases epithelial restitution by inducing Ca2+ influx after acute injury. α4 is a multiple functional protein and implicated in many aspects of cell functions by modulating protein phosphatase 2A (PP2A) stability and activity. Here we show that the clonal populations of IECs stably expressing TRPC1 (IEC-TRPC1) exhibited increased levels of α4 and PP2A catalytic subunit (PP2Ac) and that TRPC1 promoted intestinal epithelial restitution by increasing α4/PP2Ac association. The levels of α4 and PP2Ac proteins increased significantly in stable IEC-TRPC1 cells and this induction in α4/PP2Ac complexes was accompanied by an increase in IEC migration after wounding. α4 silencing by transfection with siRNA targeting α4 (siα4) or PP2Ac silencing destabilized α4/PP2Ac complexes in stable IEC-TRPC1 cells and repressed cell migration over the wounded area. Increasing the levels of cellular polyamines by stable transfection with the Odc gene stimulated α4 and PP2Ac expression and enhanced their association, thus also promoting epithelial restitution after wounding. In contrast, depletion of cellular polyamines by treatment with α-difluoromethylornithine reduced α4/PP2Ac complexes and repressed cell migration. Ectopic overexpression of α4 partially rescued rapid epithelial repair in polyamine-deficient cells. These results indicate that activation of TRPC1-mediated Ca2+ signaling enhances cell migration primarily by increasing α4/PP2Ac associations after wounding and this pathway is tightly regulated by cellular polyamines.
Collapse
Affiliation(s)
- Navneeta Rathor
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA
| | - Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA
| | - Jia-Le Song
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shelley R Wang
- Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA
| |
Collapse
|
18
|
Wang J, Tan B, Li J, Kong X, Tan M, Wu G. Regulatory role of l-proline in fetal pig growth and intestinal epithelial cell proliferation. ACTA ACUST UNITED AC 2020; 6:438-446. [PMID: 33364460 PMCID: PMC7750805 DOI: 10.1016/j.aninu.2020.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 06/14/2020] [Accepted: 07/07/2020] [Indexed: 12/27/2022]
Abstract
l-proline (Pro) is a precursor of ornithine, which is converted into polyamines via ornithine decarboxylase (ODC). Polyamines plays a key role in the proliferation of intestinal epithelial cells. The study investigated the effect of Pro on polyamine metabolism and cell proliferation on porcine enterocytes in vivo and in vitro. Twenty-four Huanjiang mini-pigs were randomly assigned into 1 of 3 groups and fed a basal diet that contained 0.77% alanine (Ala, iso-nitrogenous control), 1% Pro or 1% Pro + 0.0167% α-difluoromethylornithine (DFMO) from d 15 to 70 of gestation. The fetal body weight and number of fetuses per litter were determined, and the small and large intestines were obtained on d 70 ± 1.78 of gestation. The in vitro study was performed in intestinal porcine epithelial (IPEC-J2) cells cultured in Dulbecco's modified Eagle medium-high glucose (DMEM-H) containing 0 μmol/L Pro, 400 μmol/L Pro, or 400 μmol/L Pro + 10 mmol/L DFMO for 4 d. The results showed that maternal dietary supplementation with 1% Pro increased fetal weight; the protein and DNA concentrations of the fetal small intestine; and mRNA levels for potassium voltage-gated channel, shaker-related subfamily, member 1 (Kv1.1) in the fetal small and large intestines (P < 0.05). Supplementing Pro to either gilts or IPEC-J2 cells increased ODC protein abundances and polyamine concentrations in the fetal intestines and IPEC-J2 cells (P < 0.05). In comparison with the Pro group, the combined administration of Pro and DFMO reduced the expression of ODC protein and spermine concentration in the fetal intestine, as well as the concentrations of putrescine, spermidine and spermine in IPEC-J2 cells (P < 0.05). Meanwhile, the percentage of cells in the S-phase and the mRNA levels of proto-oncogenes c-fos and c-myc were increased in response to Pro supplementation, whereas depletion of cellular polyamines with DFMO increased tumor protein p53 (p53) mRNA levels (P < 0.05). Taken together, dietary supplementation with Pro improved fetal pig growth and intestinal epithelial cell proliferation via enhancing polyamine synthesis.
Collapse
Affiliation(s)
- Jing Wang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China.,Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Bi'e Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.,Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Jianjun Li
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Xiangfeng Kong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Minjie Tan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
19
|
Tajada S, Villalobos C. Calcium Permeable Channels in Cancer Hallmarks. Front Pharmacol 2020; 11:968. [PMID: 32733237 PMCID: PMC7358640 DOI: 10.3389/fphar.2020.00968] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer, the second cause of death worldwide, is characterized by several common criteria, known as the “cancer hallmarks” such as unrestrained cell proliferation, cell death resistance, angiogenesis, invasion and metastasis. Calcium permeable channels are proteins present in external and internal biological membranes, diffusing Ca2+ ions down their electrochemical gradient. Numerous physiological functions are mediated by calcium channels, ranging from intracellular calcium homeostasis to sensory transduction. Consequently, calcium channels play important roles in human physiology and it is not a surprise the increasing number of evidences connecting calcium channels disorders with tumor cells growth, survival and migration. Multiple studies suggest that calcium signals are augmented in various cancer cell types, contributing to cancer hallmarks. This review focuses in the role of calcium permeable channels signaling in cancer with special attention to the mechanisms behind the remodeling of the calcium signals. Transient Receptor Potential (TRP) channels and Store Operated Channels (SOC) are the main extracellular Ca2+ source in the plasma membrane of non-excitable cells, while inositol trisphosphate receptors (IP3R) are the main channels releasing Ca2+ from the endoplasmic reticulum (ER). Alterations in the function and/or expression of these calcium channels, as wells as, the calcium buffering by mitochondria affect intracellular calcium homeostasis and signaling, contributing to the transformation of normal cells into their tumor counterparts. Several compounds reported to counteract several cancer hallmarks also modulate the activity and/or the expression of these channels including non-steroidal anti-inflammatory drugs (NSAIDs) like sulindac and aspirin, and inhibitors of polyamine biosynthesis, like difluoromethylornithine (DFMO). The possible role of the calcium permeable channels targeted by these compounds in cancer and their action mechanism will be discussed also in the review.
Collapse
Affiliation(s)
- Sendoa Tajada
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Carlos Villalobos
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| |
Collapse
|
20
|
Tirtom NE, Hsu Y, Li HW. Polyamines stimulate RecA-mediated recombination by condensing duplex DNA and stabilizing intermediates. Phys Chem Chem Phys 2020; 22:11928-11935. [PMID: 32432615 DOI: 10.1039/d0cp01061h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Polyamines are naturally occurring cationic molecules in cells. In addition to their roles in modulating gene expression and cell proliferation, they have been shown to stimulate DNA recombination. The molecular mechanism for stimulation is not clear. We utilized single-molecule tethered particle motion (TPM) experiments to investigate how polyamines stimulate RecA-mediated recombination. We showed that natural polyamines, spermine and spermidine, condense duplex DNA, but with different efficiencies. While ∼300 μM of spermine condenses 50% of duplex DNA, 2.0 mM of spermidine is required to achieve the same level of condensation. The condensation takes place in a stepwise manner, and is reversible upon removal of polyamines. We also showed that addition of polyamines stimulates the duplex capture activity of RecA filament and stabilizes the intermediates with longer dwell time. Through condensing duplex DNA and stabilizing the complex of RecA filaments and duplex DNA, polyamines stimulate the formation of functional intermediates by ∼20-fold, and promote recombination progression.
Collapse
Affiliation(s)
| | - Yang Hsu
- Department of Chemistry, National Taiwan Normal University, Taiwan
| | - Hung-Wen Li
- Department of Chemistry, National Taiwan University, Taiwan.
| |
Collapse
|
21
|
TRPC1 and ORAI1 channels in colon cancer. Cell Calcium 2019; 81:59-66. [DOI: 10.1016/j.ceca.2019.06.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/24/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022]
|
22
|
Oliphant K, Allen-Vercoe E. Macronutrient metabolism by the human gut microbiome: major fermentation by-products and their impact on host health. MICROBIOME 2019; 7:91. [PMID: 31196177 PMCID: PMC6567490 DOI: 10.1186/s40168-019-0704-8] [Citation(s) in RCA: 674] [Impact Index Per Article: 134.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 05/28/2019] [Indexed: 05/11/2023]
Abstract
The human gut microbiome is a critical component of digestion, breaking down complex carbohydrates, proteins, and to a lesser extent fats that reach the lower gastrointestinal tract. This process results in a multitude of microbial metabolites that can act both locally and systemically (after being absorbed into the bloodstream). The impact of these biochemicals on human health is complex, as both potentially beneficial and potentially toxic metabolites can be yielded from such microbial pathways, and in some cases, these effects are dependent upon the metabolite concentration or organ locality. The aim of this review is to summarize our current knowledge of how macronutrient metabolism by the gut microbiome influences human health. Metabolites to be discussed include short-chain fatty acids and alcohols (mainly yielded from monosaccharides); ammonia, branched-chain fatty acids, amines, sulfur compounds, phenols, and indoles (derived from amino acids); glycerol and choline derivatives (obtained from the breakdown of lipids); and tertiary cycling of carbon dioxide and hydrogen. Key microbial taxa and related disease states will be referred to in each case, and knowledge gaps that could contribute to our understanding of overall human wellness will be identified.
Collapse
Affiliation(s)
- Kaitlyn Oliphant
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1 Canada
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1 Canada
| |
Collapse
|
23
|
TRPC-mediated Ca 2+ signaling and control of cellular functions. Semin Cell Dev Biol 2019; 94:28-39. [PMID: 30738858 DOI: 10.1016/j.semcdb.2019.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/30/2019] [Accepted: 02/06/2019] [Indexed: 12/15/2022]
Abstract
Canonical members of the TRP superfamily of ion channels have long been recognized as key elements of Ca2+ handling in a plethora of cell types. The emerging role of TRPC channels in human physiopathology has generated considerable interest in their pharmacological targeting, which requires detailed understanding of their molecular function. Although consent has been reached that receptor-phospholipase C (PLC) pathways and generation of lipid mediators constitute the prominent upstream signaling process that governs channel activity, multimodal sensing features of TRPC complexes have been demonstrated repeatedly. Downstream signaling by TRPC channels is similarly complex and involves the generation of local and global cellular Ca2+ rises, which are well-defined in space and time to govern specific cellular functions. These TRPC-mediated Ca2+ signals rely in part on Ca2+ permeation through the channels, but are essentially complemented by secondary mechanisms such as Ca2+ mobilization from storage sites and Na+/Ca2+ exchange, which involve coordinated interaction with signaling partners. Consequently, the control of cell functions by TRPC molecules is critically determined by dynamic assembly and subcellular targeting of the TRPC complexes. The very recent availability of high-resolution structure information on TRPC channel complexes has paved the way towards a comprehensive understanding of signal transduction by TRPC channels. Here, we summarize current concepts of cation permeation in TRPC complexes, TRPC-mediated shaping of cellular Ca2+ signals and the associated control of specific cell functions.
Collapse
|
24
|
Jiang LP, Wang SR, Chung HK, Buddula S, Wang JY, Rao JN. miR-222 represses expression of zipcode binding protein-1 and phospholipase C-γ1 in intestinal epithelial cells. Am J Physiol Cell Physiol 2019; 316:C415-C423. [PMID: 30649922 DOI: 10.1152/ajpcell.00165.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Both zipcode binding protein-1 (ZBP1) and phospholipase C-γ1 (PLCγ1) are intimately involved in many aspects of early intestinal mucosal repair after acute injury, but the exact mechanisms that control their cellular abundances remain largely unknown. The present study shows that microRNA-222 (miR-222) interacts with the mRNAs encoding ZBP1 and PLCγ1 and regulates ZBP1 and PLCγ1 expression in intestinal epithelial cells (IECs). The biotinylated miR-222 bound specifically to the ZBP1 and PLCγ1 mRNAs in IECs. Ectopically expressed miR-222 precursor destabilized the ZBP1 and PLCγ1 mRNAs and consequently lowered the levels of cellular ZBP1 and PLCγ1 proteins. Conversely, decreasing the levels of cellular miR-222 by transfection with its antagonism increased the stability of the ZBP1 and PLCγ1 mRNAs and increased the levels of ZBP1 and PLCγ1 proteins. Overexpression of miR-222 also inhibited cell migration over the wounded area, which was partially abolished by overexpressing ZBP1 and PLCγ1. Furthermore, prevention of the increased levels of ZBP1 and PLCγ1 in the miR-222-silenced cells by transfection with specific small interfering RNAs targeting ZBP1 or PLCγ1 mRNA inhibited cell migration after wounding. These findings indicate that induced miR-222 represses expression of ZBP1 and PLCγ1 at the posttranscriptional level, thus inhibiting IEC migration during intestinal epithelial restitution after wounding.
Collapse
Affiliation(s)
- Li-Ping Jiang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland
| | - Shelley R Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center , Baltimore, Maryland
| | - Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center , Baltimore, Maryland
| | - Saharsh Buddula
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center , Baltimore, Maryland.,Department of Pathology, University of Maryland School of Medicine , Baltimore, Maryland
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center , Baltimore, Maryland
| |
Collapse
|
25
|
Inhibition of Polyamine Biosynthesis Reverses Ca 2+ Channel Remodeling in Colon Cancer Cells. Cancers (Basel) 2019; 11:cancers11010083. [PMID: 30642111 PMCID: PMC6357118 DOI: 10.3390/cancers11010083] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/05/2019] [Accepted: 01/09/2019] [Indexed: 12/14/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) is the most important Ca2+ entry pathway in non-excitable cells. Colorectal cancer (CRC) shows decreased Ca2+ store content and enhanced SOCE that correlate with cancer hallmarks and are associated to remodeling of store-operated channels (SOCs). Normal colonic cells display small, Ca2+-selective currents driven by Orai1 channels. In contrast, CRC cells display larger, non-selective currents driven by Orai1 and transient receptor potential canonical type 1 channels (TRPC1). Difluoromethylornithine (DFMO), a suicide inhibitor of ornithine decarboxylase (ODC), the limiting step in polyamine biosynthesis, strongly prevents CRC, particularly when combined with sulindac. We asked whether DFMO may reverse SOC remodeling in CRC. We found that CRC cells overexpress ODC and treatment with DFMO decreases cancer hallmarks including enhanced cell proliferation and apoptosis resistance. Consistently, DFMO enhances Ca2+ store content and decreases SOCE in CRC cells. Moreover, DFMO abolish selectively the TRPC1-dependent component of SOCs characteristic of CRC cells and this effect is reversed by the polyamine putrescine. Combination of DFMO and sulindac inhibit both SOC components and abolish SOCE in CRC cells. Finally, DFMO treatment inhibits expression of TRPC1 and stromal interaction protein 1 (STIM1) in CRC cells. These results suggest that polyamines contribute to Ca2+ channel remodeling in CRC, and DFMO may prevent CRC by reversing channel remodeling.
Collapse
|
26
|
Zhao M, Chen Y, Wang C, Xiao W, Chen S, Zhang S, Yang L, Li Y. Systems Pharmacology Dissection of Multi-Scale Mechanisms of Action of Huo-Xiang-Zheng-Qi Formula for the Treatment of Gastrointestinal Diseases. Front Pharmacol 2019; 9:1448. [PMID: 30687082 PMCID: PMC6336928 DOI: 10.3389/fphar.2018.01448] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/26/2018] [Indexed: 12/19/2022] Open
Abstract
Multi-components Traditional Chinese Medicine (TCM) treats various complex diseases (multi-etiologies and multi-symptoms) via herbs interactions to exert curative efficacy with less adverse effects. However, the ancient Chinese compatibility theory of herbs formula still remains ambiguous. Presently, this combination principle is dissected through a systems pharmacology study on the mechanism of action of a representative TCM formula, Huo-xiang-zheng-qi (HXZQ) prescription, on the treatment of functional dyspepsia (FD), a chronic or recurrent clinical disorder of digestive system, as typical gastrointestinal (GI) diseases which burden human physical and mental health heavily and widely. In approach, a systems pharmacology platform which incorporates the pharmacokinetic and pharmaco-dynamics evaluation, target fishing and network pharmacological analyses is employed. As a result, 132 chemicals and 48 proteins are identified as active compounds and FD-related targets, and the mechanism of HXZQ formula for the treatment of GI diseases is based on its three function modules of anti-inflammation, immune protection and gastrointestinal motility regulation mainly through four, i.e., PIK-AKT, JAK-STAT, Toll-like as well as Calcium signaling pathways. In addition, HXZQ formula conforms to the ancient compatibility rule of "Jun-Chen-Zuo-Shi" due to the different, while cooperative roles that herbs possess, specifically, the direct FD curative effects of GHX (serving as Jun drug), the anti-bacterial efficacy and major accompanying symptoms-reliving bioactivities of ZS and BZ (as Chen), the detoxication and ADME regulation capacities of GC (as Shi), as well as the minor symptoms-treating efficacy of the rest 7 herbs (as Zuo). This work not only provides an insight of the therapeutic mechanism of TCMs on treating GI diseases from a multi-scale perspective, but also may offer an efficient way for drug discovery and development from herbal medicine as complementary drugs.
Collapse
Affiliation(s)
- Miaoqing Zhao
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian, China.,Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Shihezi, China
| | - Yangyang Chen
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, China
| | - Chao Wang
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian, China
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Shusheng Chen
- Systems Biology Laboratory, Department of Computer & Information Science & Engineering, University of Florida, Gainesville, FL, United States
| | - Shuwei Zhang
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian, China
| | - Ling Yang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Li
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian, China.,Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Shihezi, China
| |
Collapse
|
27
|
Abstract
Advances in our understanding of the metabolism and molecular functions of polyamines and their alterations in cancer have led to resurgence in the interest of targeting polyamine metabolism as an anticancer strategy. Increasing knowledge of the interplay between polyamine metabolism and other cancer-driving pathways, including the PTEN-PI3K-mTOR complex 1 (mTORC1), WNT signalling and RAS pathways, suggests potential combination therapies that will have considerable clinical promise. Additionally, an expanding number of promising clinical trials with agents targeting polyamines for both therapy and prevention are ongoing. New insights into molecular mechanisms linking dysregulated polyamine catabolism and carcinogenesis suggest additional strategies that can be used for cancer prevention in at-risk individuals. In addition, polyamine blocking therapy, a strategy that combines the inhibition of polyamine biosynthesis with the simultaneous blockade of polyamine transport, can be more effective than therapies based on polyamine depletion alone and may involve an antitumour immune response. These findings open up new avenues of research into exploiting aberrant polyamine metabolism for anticancer therapy.
Collapse
Affiliation(s)
- Robert A Casero
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| | - Tracy Murray Stewart
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Anthony E Pegg
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
28
|
Polysaccharide extracts of Astragalus membranaceus and Atractylodes macrocephala promote intestinal epithelial cell migration by activating the polyamine-mediated K+ channel. Chin J Nat Med 2018; 16:674-682. [DOI: 10.1016/s1875-5364(18)30107-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Indexed: 12/22/2022]
|
29
|
Rathor N, Chung HK, Wang SR, Qian M, Turner DJ, Wang JY, Rao JN. β-PIX plays an important role in regulation of intestinal epithelial restitution by interacting with GIT1 and Rac1 after wounding. Am J Physiol Gastrointest Liver Physiol 2018; 314:G399-G407. [PMID: 29191942 PMCID: PMC5899242 DOI: 10.1152/ajpgi.00296.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Early gut mucosal restitution is a process by which intestinal epithelial cells (IECs) migrate over the wounded area, and its defective regulation occurs commonly in various critical pathological conditions. This rapid reepithelialization is mediated by different activating small GTP-binding proteins, but the exact mechanism underlying this process remains largely unknown. Recently, it has been reported that interaction between p21-activated kinase-interacting exchange factor (β-PIX) and G protein-coupled receptor kinase-interacting protein 1 (GIT1) activates small GTPases and plays an important role in the regulation of cell motility. Here, we show that induced association of β-PIX with GIT1 is essential for the stimulation of IEC migration after wounding by activating Rac1. Levels of β-PIX and GIT1 proteins and their association in differentiated IECs (line of IEC-Cdx2L1) were much higher than those observed in undifferentiated IECs (line of IEC-6), which was associated with an increase in IEC migration after wounding. Decreased levels of endogenous β-PIX by its gene-silencing destabilized β-PIX/GIT1 complexes, repressed Rac1 activity and inhibited cell migration over the wounded area. In contrast, ectopic overexpression of β-PIX increased the levels of β-PIX/GIT1 complexes, stimulated Rac1 activity, and enhanced intestinal epithelial restitution. Increased levels of cellular polyamines also stimulated β-PIX/GIT1 association, increased Rac1 activity, and promoted the epithelial restitution. Moreover, polyamine depletion decreased cellular abundances of β-PIX/GIT1 complex and repressed IEC migration after wounding, which was rescued by ectopic overexpression of β-PIX or GIT1. These results indicate that β-PIX/GIT1/Rac1 association is necessary for stimulation of IEC migration after wounding and that this signaling pathway is tightly regulated by cellular polyamines. NEW & NOTEWORTHY Our current study demonstrates that induced association of β-PIX with GIT1 is essential for the stimulation of intestinal epithelial restitution by activating Rac1, and this signaling pathway is tightly regulated by cellular polyamines.
Collapse
Affiliation(s)
- Navneeta Rathor
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Hee Kyoung Chung
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Shelley R. Wang
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Michael Qian
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland
| | - Douglas J. Turner
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Jian-Ying Wang
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland,3Department of Pathology, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jaladanki N. Rao
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| |
Collapse
|
30
|
Abu-Khudir R, Habieb ME, Mohamed MA, Hawas AM, Mohamed TM. Anti-apoptotic role of spermine against lead and/or gamma irradiation-induced hepatotoxicity in male rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:24272-24283. [PMID: 28889190 DOI: 10.1007/s11356-017-0069-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 09/01/2017] [Indexed: 06/07/2023]
Abstract
Exposure to either lead (Pb) or γ-irradiation (IR) results in oxidative stress in biological systems. Herein, we explored the potential anti-apoptotic effect of spermine (Spm) against lead and/or γ-irradiation-induced hepatotoxicity in male albino rats. Rats were divided into eight experimental groups of ten rats each: groups including negative control, whole body γ-irradiated (6 Gray (Gy)), lead acetate (PbAct) trihydrate orally administered (75 mg/kg bw ≡ 40 mg/kg bw Pb for 14 consecutive days), and Spm intraperitoneally dosed (10 mg/kg bw for 14 consecutive days) rats and groups subjected to combinations of Pb + IR, Spm + IR, Spm + Pb, and Spm + Pb followed by IR on day 14 (Spm + Pb + IR). A significant decrease in arginase activity as well as mRNA and protein levels of Bcl-2 and p21 was observed in rats intoxicated with Pb and/or γ-irradiation compared to controls, whereas Bax mRNA and protein levels were significantly increased. Also, an increased level of nitric oxide (NO) with a reduced arginase activity was observed in liver tissues of intoxicated rats. Spm co-treatment with lead and/or γ-irradiation attenuated the increase in Bax mRNA and protein expression, while it restored those of Bcl-2 and p21 together with NO levels and arginase activity to control values. Altogether, we suggest that Spm may be useful in combating free radical-induced apoptosis in Pb-intoxicated and/or γ-irradiated rats.
Collapse
Affiliation(s)
- Rasha Abu-Khudir
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, P.O. 31527, Tanta, Egypt.
| | - Mahmoud E Habieb
- Drug Radiation Research Department, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, P.O. Box; 29, Nasr City, Cairo, Egypt
| | - Marwa A Mohamed
- Drug Radiation Research Department, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, P.O. Box; 29, Nasr City, Cairo, Egypt
| | - Asrar M Hawas
- Drug Radiation Research Department, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, P.O. Box; 29, Nasr City, Cairo, Egypt
| | - Tarek M Mohamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, P.O. 31527, Tanta, Egypt
| |
Collapse
|
31
|
Gu M, Bai N, Xu B, Xu X, Jia Q, Zhang Z. Protective effect of glutamine and arginine against soybean meal-induced enteritis in the juvenile turbot (Scophthalmus maximus). FISH & SHELLFISH IMMUNOLOGY 2017; 70:95-105. [PMID: 28882796 DOI: 10.1016/j.fsi.2017.08.048] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/13/2017] [Accepted: 08/25/2017] [Indexed: 06/07/2023]
Abstract
Soybean meal can induce enteritis in the distal intestine (DI) and decrease the immunity of several cultured fish species, including turbot Scophthalmus maximus. Glutamine and arginine supplementation have been used to improve immunity and intestinal morphology in fish. This study was conducted to investigate the effects of these two amino acids on the immunity and intestinal health of turbot suffering from soybean meal-induced enteritis. Turbots (initial weight 7.6 g) were fed one of three isonitrogenous and isolipidic diets for 8 weeks: SBM (control diet), with 40% soybean meal; GLN, SBM diet plus 1.5% glutamine; ARG, the SBM diet plus 1.5% arginine. Symptoms that are typical of soybean meal-induced enteritis, including swelling of the lamina propria and subepithelial mucosa and a strong infiltration of various inflammatory cells was observed in fish that fed the SBM diet. Glutamine and arginine supplementation significantly increased (1) the weight gain and feed efficiency ratio; (2) the height and vacuolization of villi and the integrity of microvilli in DI; (3) serum lysozyme activity, and the concentrations of C3, C4, and IgM. These two amino acids also significantly decreased the infiltration of leucocytes in the lamina propria and submucosa and the expression of inflammatory cytokines including il-8, tnf-α, and tgf-β. For the mucosal microbiota, arginine supplementation significantly increased microbiota community richness and diversity, and glutamine supplementation significantly increased the relative abundance of Lactobacillus and Bacillus. These results indicate that dietary glutamine and arginine improved the growth performance, feed utilization, and distal intestinal morphology, activated the innate and adaptive immune systems, changed the intestinal mucosal microbiota community, and relieved SBMIE possibly by suppression of the inflammation response.
Collapse
Affiliation(s)
- Min Gu
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China
| | - Nan Bai
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China.
| | - Bingying Xu
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China
| | - Xiaojie Xu
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China
| | - Qian Jia
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China
| | - Zhiyu Zhang
- Marine College, Shandong University at Weihai, 180 Wenhua West Road, Weihai, 264209, PR China
| |
Collapse
|
32
|
Wang PY, Wang SR, Xiao L, Chen J, Wang JY, Rao JN. c-Jun enhances intestinal epithelial restitution after wounding by increasing phospholipase C-γ1 transcription. Am J Physiol Cell Physiol 2017; 312:C367-C375. [PMID: 28100486 DOI: 10.1152/ajpcell.00330.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/10/2017] [Accepted: 01/10/2017] [Indexed: 01/06/2023]
Abstract
c-Jun is an activating protein 1 (AP-1) transcription factor and implicated in many aspects of cellular functions, but its exact role in the regulation of early intestinal epithelial restitution after injury remains largely unknown. Phospholipase C-γ1 (PLCγ1) catalyzes hydrolysis of phosphatidylinositol 4,5 biphosphate into the second messenger diacylglycerol and inositol 1,4,5 triphosphate, coordinates Ca2+ store mobilization, and regulates cell migration and proliferation in response to stress. Here we reported that c-Jun upregulates PLCγ1 expression and enhances PLCγ1-induced Ca2+ signaling, thus promoting intestinal epithelial restitution after wounding. Ectopically expressed c-Jun increased PLCγ1 expression at the transcription level, and this stimulation is mediated by directly interacting with AP-1 and CCAAT-enhancer-binding protein (C/EBP) binding sites that are located at the proximal region of the rat PLCγ1 promoter. Increased levels of PLCγ1 by c-Jun elevated cytosolic free Ca2+ concentration and stimulated intestinal epithelial cell migration over the denuded area after wounding. The c-Jun-mediated PLCγ1/Ca2+ signal also plays an important role in polyamine-induced cell migration after wounding because increased c-Jun rescued Ca2+ influx and cell migration in polyamine-deficient cells. These findings indicate that c-Jun induces PLCγ1 expression transcriptionally and enhances rapid epithelial restitution after injury by activating Ca2+ signal.
Collapse
Affiliation(s)
- Peng-Yuan Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Shelley R Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Jie Chen
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland; and.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; .,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| |
Collapse
|
33
|
STIM-TRP Pathways and Microdomain Organization: Ca 2+ Influx Channels: The Orai-STIM1-TRPC Complexes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:139-157. [PMID: 28900913 DOI: 10.1007/978-3-319-57732-6_8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ca2+ influx by plasma membrane Ca2+ channels is the crucial component of the receptor-evoked Ca2+ signal. The two main Ca2+ influx channels of non-excitable cells are the Orai and TRPC families of Ca2+ channels. These channels are activated in response to cell stimulation and Ca2+ release from the endoplasmic reticulum (ER). The protein that conveys the Ca2+ content of the ER to the plasma membrane is the ER Ca2+ sensor STIM1. STIM1 activates the Orai channels and is obligatory for channel opening. TRPC channels can function in two modes, as STIM1-dependent and STIM1-independent. When activated by STIM1, both channel types function at the ER/PM (plasma membrane) junctions. This chapter describes the properties and regulation of the channels by STIM1, with emphasis how and when TRPC channels function as STIM1-dependent and STIM1-independent modes and their unique Ca2+-dependent physiological functions that are not shared with the Orai channels.
Collapse
|
34
|
STIM-TRP Pathways and Microdomain Organization: Contribution of TRPC1 in Store-Operated Ca 2+ Entry: Impact on Ca 2+ Signaling and Cell Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:159-188. [PMID: 28900914 DOI: 10.1007/978-3-319-57732-6_9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Store-operated calcium entry (SOCE) is a ubiquitous Ca2+ entry pathway that is activated in response to depletion of ER-Ca2+ stores and critically controls the regulation of physiological functions in a wide variety of cell types. The transient receptor potential canonical (TRPC) channels (TRPCs 1-7), which are activated by stimuli leading to PIP2 hydrolysis, were first identified as molecular components of SOCE channels. While TRPC1 was associated with SOCE and regulation of function in several cell types, none of the TRPC members displayed I CRAC, the store-operated current identified in lymphocytes and mast cells. Intensive search finally led to the identification of Orai1 and STIM1 as the primary components of the CRAC channel. Orai1 was established as the pore-forming channel protein and STIM1 as the ER-Ca2+ sensor protein involved in activation of Orai1. STIM1 also activates TRPC1 via a distinct domain in its C-terminus. However, TRPC1 function depends on Orai1-mediated Ca2+ entry, which triggers recruitment of TRPC1 into the plasma membrane where it is activated by STIM1. TRPC1 and Orai1 form distinct store-operated Ca2+ channels that regulate specific cellular functions. It is now clearly established that regulation of TRPC1 trafficking can change plasma membrane levels of the channel, the phenotype of the store-operated Ca2+ current, as well as pattern of SOCE-mediated [Ca2+]i signals. Thus, TRPC1 is activated downstream of Orai1 and modifies the initial [Ca2+]i signal generated by Orai1. This review will highlight current concepts of the activation and regulation of TRPC1 channels and its impact on cell function.
Collapse
|
35
|
Wu X, Cao W, Jia G, Zhao H, Chen X, Wu C, Tang J, Wang J, Liu G. New insights into the role of spermine in enhancing the antioxidant capacity of rat spleen and liver under oxidative stress. ACTA ACUST UNITED AC 2016; 3:85-90. [PMID: 29767047 PMCID: PMC5941080 DOI: 10.1016/j.aninu.2016.11.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/14/2016] [Indexed: 11/24/2022]
Abstract
Oxidative stress can damage cellular antioxidant defense and reduce livestock production efficiency. Spermine is a ubiquitous cellular component that plays important roles in stabilizing nucleic acids, modulating cell growth and differentiation, and regulating ion channel activities. Spermine has the potential to alleviate the effects of oxidative stress. However, to date no information is available about the effect of spermine administration on antioxidant property of the liver and spleen in any mammalian in vivo system. This study aims to investigate the protective effect of spermine on rat liver and spleen under oxidative stress. Rats received intragastric administration of either 0.4 μmol/g body weight of spermine or saline once a day for 3 days. The rats in each treatment were then injected with either diquat or sterile saline at 12 mg/kg body weight. Liver and spleen samples were collected 48 h after the last spermine ingestion. Results showed that regardless of diquat treatment, spermine administration significantly reduced the malondialdehyde (MDA) content by 23.78% in the liver and by 5.75% in the spleen, respectively (P < 0.05). Spermine administration also enhanced the catalase (CAT) activity, anti-hydroxyl radical (AHR) capacity and glutathione (GSH) content by 38.68%, 15.53% and 1.32% in the spleen, respectively (P < 0.05). There were interactions between spermine administration and diquat injection about anti-superoxide anion (ASA), AHR capacity, CAT activity, GSH content, and total antioxidant capacity (T-AOC) in the liver and about ASA capacity and T-AOC in the spleen of weaned rats (P < 0.05). Compared with the control group, spermine administration significantly increased the AHR capacity, CAT activity, GSH content, and T-AOC by 40.23%, 31.15%, 30.25%, 35.37% in the liver, respectively (P < 0.05) and increased the T-AOC by 8% in the spleen of weaned rats (P < 0.05). Compared with the diquat group, spermine + diquat group significantly increased ASA capacity by 15.63% in the liver and by 73.41% in the spleen of weaned rats, respectively (P < 0.05). Results demonstrate that spermine administration can increase the antioxidant capacity in the liver and spleen and can enhance the antioxidant status in the spleen and liver under oxidative stress.
Collapse
Affiliation(s)
- Xianjian Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, China
| | - Wei Cao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, China
| | - Gang Jia
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, China
| | - Hua Zhao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, China
| | - Caimei Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, China
| | - Jiayong Tang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, China
| | - Jing Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Guangmang Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, China
- Corresponding author.
| |
Collapse
|
36
|
Abstract
The content of spermidine and spermine in mammalian cells has important roles in protein and nucleic acid synthesis and structure, protection from oxidative damage, activity of ion channels, cell proliferation, differentiation, and apoptosis. Spermidine is essential for viability and acts as the precursor of hypusine, a post-translational addition to eIF5A allowing the translation of mRNAs encoding proteins containing polyproline tracts. Studies with Gy mice and human patients with the very rare X-linked genetic condition Snyder-Robinson syndrome that both lack spermine synthase show clearly that the correct spermine:spermidine ratio is critical for normal growth and development.
Collapse
Affiliation(s)
- Anthony E Pegg
- From the Department of Cellular and Molecular Physiology, Milton S. Hershey Medical Center, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| |
Collapse
|
37
|
Wang J, Li GR, Tan BE, Xiong X, Kong XF, Xiao DF, Xu LW, Wu MM, Huang B, Kim SW, Yin YL. Oral administration of putrescine and proline during the suckling period improves epithelial restitution after early weaning in piglets. J Anim Sci 2016; 93:1679-88. [PMID: 26020189 DOI: 10.2527/jas.2014-8230] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Polyamines are necessary for normal integrity and the restitution after injury of the gastrointestinal epithelium. The objective of this study was to investigate the effects of oral administration of putrescine and proline during the suckling period on epithelial restitution after early weaning in piglets. Eighteen neonatal piglets (Duroc × Landrace × Large Yorkshire) from 3 litters (6 piglets per litter) were assigned to 3 groups, representing oral administration with an equal volume of saline (control), putrescine (5 mg/kg BW), and proline (25 mg/kg BW) twice daily from d 1 to weaning at 14 d of age. Plasma and intestinal samples were obtained 3 d after weaning. The results showed that oral administration of putrescine or proline increased the final BW and ADG of piglets compared with the control (P < 0.05). Proline treatment decreased plasma D-lactate concentration but increased the villus height in the jejunum and ileum, as well as the percentage of proliferating cell nuclear antigen (PCNA) positive cells and alkaline phosphatase (AKP) activity in the jejunal mucosa (P < 0.05). The protein expressions for zonula occludens (ZO-1), occludin, and claudin-3 (P < 0.05) but not mRNA were increased in the jejunum of putrescine- and proline-treated piglets compared with those of control piglets. The voltage-gated K+ channel (Kv) 1.1 protein expression in the jejunum of piglets administrated with putrescine and the Kv1.5 mRNA and Kv1.1 protein levels in the ileum of piglets administrated with proline were greater than those in control piglets (P < 0.05). These findings indicate that polyamine or its precursor could improve mucosal proliferation, intestinal morphology, as well as tight junction and potassium channel protein expressions in early-weaned piglets, with implications for epithelial restitution and barrier function after stress injury.
Collapse
|
38
|
Ong HL, de Souza LB, Ambudkar IS. Role of TRPC Channels in Store-Operated Calcium Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:87-109. [DOI: 10.1007/978-3-319-26974-0_5] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
39
|
Chung HK, Rathor N, Wang SR, Wang JY, Rao JN. RhoA enhances store-operated Ca2+ entry and intestinal epithelial restitution by interacting with TRPC1 after wounding. Am J Physiol Gastrointest Liver Physiol 2015; 309:G759-67. [PMID: 26336927 PMCID: PMC4628965 DOI: 10.1152/ajpgi.00185.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/26/2015] [Indexed: 01/31/2023]
Abstract
Early mucosal restitution occurs as a consequence of epithelial cell migration to resealing of superficial wounds after injury. Our previous studies show that canonical transient receptor potential-1 (TRPC1) functions as a store-operated Ca(2+) channel (SOC) in intestinal epithelial cells (IECs) and plays an important role in early epithelial restitution by increasing Ca(2+) influx. Here we further reported that RhoA, a small GTP-binding protein, interacts with and regulates TRPC1, thus enhancing SOC-mediated Ca(2+) entry (SOCE) and epithelial restitution after wounding. RhoA physically associated with TRPC1 and formed the RhoA/TRPC1 complexes, and this interaction increased in stable TRPC1-transfected IEC-6 cells (IEC-TRPC1). Inactivation of RhoA by treating IEC-TRPC1 cells with exoenzyme C3 transferase (C3) or ectopic expression of dominant negative RhoA (DNMRhoA) reduced RhoA/TRPC1 complexes and inhibited Ca(2+) influx after store depletion, which was paralleled by an inhibition of cell migration over the wounded area. In contrast, ectopic expression of wild-type (WT)-RhoA increased the levels of RhoA/TRPC1 complexes, induced Ca(2+) influx through activation of SOCE, and promoted cell migration after wounding. TRPC1 silencing by transfecting stable WT RhoA-transfected cells with siRNA targeting TRPC1 (siTRPC1) reduced SOCE and repressed epithelial restitution. Moreover, ectopic overexpression of WT-RhoA in polyamine-deficient cells rescued the inhibition of Ca(2+) influx and cell migration induced by polyamine depletion. These findings indicate that RhoA interacts with and activates TRPC1 and thus stimulates rapid epithelial restitution after injury by inducing Ca(2+) signaling.
Collapse
Affiliation(s)
- Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Navneeta Rathor
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Shelley R Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland; and Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| |
Collapse
|
40
|
Ong HL, Ambudkar IS. Molecular determinants of TRPC1 regulation within ER–PM junctions. Cell Calcium 2015; 58:376-86. [DOI: 10.1016/j.ceca.2015.03.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 11/30/2022]
|
41
|
Ca2+ signaling in cytoskeletal reorganization, cell migration, and cancer metastasis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:409245. [PMID: 25977921 PMCID: PMC4421034 DOI: 10.1155/2015/409245] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/12/2015] [Indexed: 01/19/2023]
Abstract
Proper control of Ca2+ signaling is mandatory for effective cell migration, which is critical for embryonic development, wound healing, and cancer metastasis. However, how Ca2+ coordinates structural components and signaling molecules for proper cell motility had remained elusive. With the advance of fluorescent live-cell Ca2+ imaging in recent years, we gradually understand how Ca2+ is regulated spatially and temporally in migrating cells, driving polarization, protrusion, retraction, and adhesion at the right place and right time. Here we give an overview about how cells create local Ca2+ pulses near the leading edge, maintain cytosolic Ca2+ gradient from back to front, and restore Ca2+ depletion for persistent cell motility. Differential roles of Ca2+ in regulating various effectors and the interaction of roles of Ca2+ signaling with other pathways during migration are also discussed. Such information might suggest a new direction to control cancer metastasis by manipulating Ca2+ and its associating signaling molecules in a judicious manner.
Collapse
|
42
|
Abstract
Advanced mucosal healing (MH) after intestinal mucosal inflammation coincides with sustained clinical remission and reduced rates of hospitalization and surgical resection, explaining why MH is increasingly considered as a full therapeutic goal and as an endpoint for clinical trials. Intestinal MH is a complex phenomenon viewed as a succession of steps necessary to restore tissue structure and function. These steps include epithelial cell migration and proliferation, cell differentiation, restoration of epithelial barrier functions, and modulation of cell apoptosis. Few clinical studies have evaluated the needs for specific macronutrients and micronutrients and their effects on intestinal MH, most data having been obtained from animal and cell studies. These data suggest that supplementation with specific amino acids including arginine, glutamine, glutamate, threonine, methionine, serine, proline, and the amino acid-derived compounds, polyamines can favorably influence MH. Short-chain fatty acids, which are produced by the microbiota from undigested polysaccharides and protein-derived amino acids, also exert beneficial effects on the process of intestinal MH in experimental models. Regarding supplementation with lipids, although the effects of ω-3 and ω-6 fatty acids remain controversial, endogenous prostaglandin synthesis seems to be necessary for MH. Finally, among micronutrients, several vitamin and mineral deficiencies with different frequencies have been observed in patients with inflammatory bowel diseases and supplementation with some of them (vitamin A, vitamin D3, vitamin C, and zinc) are presumed to favor MH. Future work, including clinical studies, should evaluate the efficiency of supplementation with combination of dietary compounds as adjuvant nutritional intervention for MH of the inflamed intestinal mucosa.
Collapse
|
43
|
Shalygin A, Skopin A, Kalinina V, Zimina O, Glushankova L, Mozhayeva GN, Kaznacheyeva E. STIM1 and STIM2 proteins differently regulate endogenous store-operated channels in HEK293 cells. J Biol Chem 2014; 290:4717-4727. [PMID: 25533457 DOI: 10.1074/jbc.m114.601856] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The endoplasmic reticulum calcium sensors stromal interaction molecules 1 and 2 (STIM1 and STIM2) are key modulators of store-operated calcium entry. Both these sensors play a major role in physiological functions in normal tissue and in pathology, but available data on native STIM2-regulated plasma membrane channels are scarce. Only a few studies have recorded STIM2-induced CRAC (calcium release-activated calcium) currents. On the other hand, many cell types display store-operated currents different from CRAC. The STIM1 protein regulates not only CRAC but also transient receptor potential canonical (TRPC) channels, but it has remained unclear whether STIM2 is capable of regulating store-operated non-CRAC channels. Here we present for the first time experimental evidence for the existence of endogenous non-CRAC STIM2-regulated channels. As shown in single-channel patch clamp experiments on HEK293 cells, selective activation of native STIM2 proteins or STIM2 overexpression results in store-operated activation of Imin channels, whereas STIM1 activation blocks this process. Changes in the ratio between active STIM2 and STIM1 proteins can switch the regulation of Imin channels between store-operated and store-independent modes. We have previously characterized electrophysiological properties of different Ca(2+) influx channels coexisting in HEK293 cells. The results of this study show that STIM1 and STIM2 differ in the ability to activate these store-operated channels; Imin channels are regulated by STIM2, TRPC3-containing INS channels are induced by STIM1, and TRPC1-composed Imax channels are activated by both STIM1 and STIM2. These new data about cross-talk between STIM1 and STIM2 and their different roles in store-operated channel activation are indicative of an additional level in the regulation of store-operated calcium entry pathways.
Collapse
Affiliation(s)
- Alexey Shalygin
- From the Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia.
| | - Anton Skopin
- From the Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - Vera Kalinina
- From the Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - Olga Zimina
- From the Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - Lyuba Glushankova
- From the Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - Galina N Mozhayeva
- From the Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - Elena Kaznacheyeva
- From the Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia.
| |
Collapse
|
44
|
Abstract
Maximizing metabolic stress at a given level of mechanical stress can improve the adaptive response to endurance training, decrease injury, and potentially improve performance. Calcium and metabolic stress, in the form of heat, decreases in the adenosine triphosphate/adenosine diphosphate ratio, glycogen depletion, caloric restriction, and oxidative stress, are the primary determinants of the adaptation to training. These stressors increase the activity and amount of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α), a protein that can directly induce the primary adaptive responses to endurance exercise: mitochondrial biogenesis, angiogenesis, and increases in fat oxidation. The activity of PGC-1α is regulated by its charge (phosphorylation and acetylation), whereas its transcription is regulated by proteins that bind to myocyte enhancing factor 2, enhancer box, and cyclic adenosine monophosphate response element sites within the PGC-1α promoter. This brief review will describe what is known about the control of PGC-1α by these metabolic stressors. As the duration of calcium release and the amount of metabolic stress, and therefore the activation of PGC-1α, can be directly modulated by training and nutrition, a simple strategy can be generated to maximize the adaptive response to endurance training.
Collapse
|
45
|
Rathor N, Chung HK, Wang SR, Wang JY, Turner DJ, Rao JN. Caveolin-1 enhances rapid mucosal restitution by activating TRPC1-mediated Ca2+ signaling. Physiol Rep 2014; 2:2/11/e12193. [PMID: 25367694 PMCID: PMC4255804 DOI: 10.14814/phy2.12193] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Early rapid mucosal restitution occurs as a consequence of epithelial cell migration to reseal superficial wounds, a process independent of cell proliferation. Our previous studies revealed that the canonical transient receptor potential-1 (TRPC1) functions as a store-operated Ca(2+) channel (SOCs) in intestinal epithelial cells (IECs) and regulates epithelial restitution after wounding, but the exact mechanism underlying TRPC1 activation remains elusive. Caveolin-1 (Cav1) is a major component protein that is associated with caveolar lipid rafts in the plasma membrane and was recently identified as a regulator of store-operated Ca(2+) entry (SOCE). Here, we showed that Cav1 plays an important role in the regulation of mucosal restitution by activating TRPC1-mediated Ca(2+) signaling. Target deletion of Cav1 delayed gastric mucosal repair after exposure to hypertonic NaCl in mice, although it did not affect total levels of TRPC1 protein. In cultured IECs, Cav1 directly interacted with TRPC1 and formed Cav1/TRPC1 complex as measured by immunoprecipitation assays. Cav1 silencing in stable TRPC1-transfected cells by transfection with siCav1 reduced SOCE without effect on the level of resting [Ca(2+)]cyt. Inhibition of Cav1 expression by siCav1 and subsequent decrease in Ca(2+) influx repressed epithelial restitution, as indicated by a decrease in cell migration over the wounded area, whereas stable ectopic overexpression of Cav1 increased Cav1/TRPC1 complex, induced SOCE, and enhanced cell migration after wounding. These results indicate that Cav1 physically interacts with and activates TRPC1, thus stimulating TRPC1-mediated Ca(2+) signaling and rapid mucosal restitution after injury.
Collapse
Affiliation(s)
- Navneeta Rathor
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland, USA Baltimore VA Medical Center, Baltimore, Maryland, USA
| | - Hee K Chung
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland, USA Baltimore VA Medical Center, Baltimore, Maryland, USA
| | - Shelley R Wang
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jian-Ying Wang
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland, USA Baltimore VA Medical Center, Baltimore, Maryland, USA Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Douglas J Turner
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland, USA Baltimore VA Medical Center, Baltimore, Maryland, USA
| | - Jaladanki N Rao
- Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland, USA Baltimore VA Medical Center, Baltimore, Maryland, USA
| |
Collapse
|
46
|
Sobradillo D, Hernández-Morales M, Ubierna D, Moyer MP, Núñez L, Villalobos C. A reciprocal shift in transient receptor potential channel 1 (TRPC1) and stromal interaction molecule 2 (STIM2) contributes to Ca2+ remodeling and cancer hallmarks in colorectal carcinoma cells. J Biol Chem 2014; 289:28765-82. [PMID: 25143380 DOI: 10.1074/jbc.m114.581678] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We have investigated the molecular basis of intracellular Ca(2+) handling in human colon carcinoma cells (HT29) versus normal human mucosa cells (NCM460) and its contribution to cancer features. We found that Ca(2+) stores in colon carcinoma cells are partially depleted relative to normal cells. However, resting Ca(2+) levels, agonist-induced Ca(2+) increases, store-operated Ca(2+) entry (SOCE), and store-operated currents (ISOC) are largely enhanced in tumor cells. Enhanced SOCE and depleted Ca(2+) stores correlate with increased cell proliferation, invasion, and survival characteristic of tumor cells. Normal mucosa cells displayed small, inward Ca(2+) release-activated Ca(2+) currents (ICRAC) mediated by ORAI1. In contrast, colon carcinoma cells showed mixed currents composed of enhanced ICRAC plus a nonselective ISOC mediated by TRPC1. Tumor cells display increased expression of TRPC1, ORAI1, ORAI2, ORAI3, and STIM1. In contrast, STIM2 protein was nearly depleted in tumor cells. Silencing data suggest that enhanced ORAI1 and TRPC1 contribute to enhanced SOCE and differential store-operated currents in tumor cells, whereas ORAI2 and -3 are seemingly less important. In addition, STIM2 knockdown decreases SOCE and Ca(2+) store content in normal cells while promoting apoptosis resistance. These data suggest that loss of STIM2 may underlie Ca(2+) store depletion and apoptosis resistance in tumor cells. We conclude that a reciprocal shift in TRPC1 and STIM2 contributes to Ca(2+) remodeling and tumor features in colon cancer.
Collapse
Affiliation(s)
- Diego Sobradillo
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Miriam Hernández-Morales
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Daniel Ubierna
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | | | - Lucía Núñez
- the Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, 47003 Valladolid, Spain
| | - Carlos Villalobos
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain,
| |
Collapse
|
47
|
Aihara E, Montrose MH. Importance of Ca(2+) in gastric epithelial restitution-new views revealed by real-time in vivo measurements. Curr Opin Pharmacol 2014; 19:76-83. [PMID: 25108560 DOI: 10.1016/j.coph.2014.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 07/22/2014] [Accepted: 07/22/2014] [Indexed: 12/14/2022]
Abstract
It has been a few decades since Ca(2+) was identified as one of the important factors that can accelerate gastric wound repair as well as contribute to epithelial homeostasis and regulation of gastric secretions. The mechanistic basis has remained largely unexplored in vivo because it was not possible to track in real time either intracellular Ca(2+) mobilization or wound repair in living tissues. Recent advances in technology, such as combining high resolution light microscopy and genetically encoded Ca(2+) reporters in mice, now allow the monitoring of Ca(2+) mobilization during gastric epithelial cell restitution. Ca(2+) is a ubiquitous second messenger that influences numerous cellular processes, including gastric acid/bicarbonate secretion, mucus secretion, and cell migration. We have demonstrated that cytosolic Ca(2+) mobilization within the restituting gastric epithelial cells is a central signal driving small wound repair. However, extracellular Ca(2+) is also mobilized in the juxtamucosal luminal space above a wound, and evidence suggests extracellular Ca(2+) is a third messenger that also promotes gastric epithelial restitution. Interplay between intracellular and extracellular Ca(2+) is necessary for efficient gastric epithelial restitution.
Collapse
Affiliation(s)
- Eitaro Aihara
- Department of Molecular and Cellular Physiology, University of Cincinnati, OH 45267, USA
| | - Marshall H Montrose
- Department of Molecular and Cellular Physiology, University of Cincinnati, OH 45267, USA.
| |
Collapse
|
48
|
Rathor N, Zhuang R, Wang JY, Donahue JM, Turner DJ, Rao JN. Src-mediated caveolin-1 phosphorylation regulates intestinal epithelial restitution by altering Ca(2+) influx after wounding. Am J Physiol Gastrointest Liver Physiol 2014; 306:G650-8. [PMID: 24557763 PMCID: PMC3989706 DOI: 10.1152/ajpgi.00003.2014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Early mucosal restitution occurs as a consequence of intestinal epithelial cell (IEC) migration to reseal superficial wounds, but its exact mechanism remains largely unknown. Caveolin-1 (Cav1), a major component associated with caveolar lipid rafts in the plasma membrane, is implicated in many aspects of cellular functions. This study determined if c-Src kinase (Src)-induced Cav1 phosphorylation promotes intestinal epithelial restitution after wounding by activating Cav1-mediated Ca(2+) signaling. Src directly interacted with Cav1, formed Cav1-Src complexes, and phosphorylated Cav1 in IECs. Inhibition of Src activity by its chemical inhibitor PP2 or suppression of the functional caveolin scaffolding domain by caveolin-scaffolding domain peptides prevented Cav1-Src interaction, reduced Cav1 phosphorylation, decreased Ca(2+) influx, and inhibited cell migration after wounding. Disruption of caveolar lipid raft microdomains by methyl-β-cyclodextrin reduced Cav1-mediated Ca(2+) influx and repressed epithelial restitution. Moreover, Src silencing prevented subcellular redistribution of phosphorylated Cav1 in migrating IECs. These results indicate that Src-induced Cav1 phosphorylation stimulates epithelial restitution by increasing Cav1-mediated Ca(2+) signaling after wounding, thus contributing to the maintenance of gut mucosal integrity under various pathological conditions.
Collapse
Affiliation(s)
- Navneeta Rathor
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and
| | - Ran Zhuang
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and
| | - Jian-Ying Wang
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and ,3Department of Pathology, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland
| | - James M. Donahue
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and
| | - Douglas J. Turner
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and
| | - Jaladanki N. Rao
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and
| |
Collapse
|
49
|
Yang H, Rao JN, Wang JY. Posttranscriptional Regulation of Intestinal Epithelial Tight Junction Barrier by RNA-binding Proteins and microRNAs. Tissue Barriers 2014; 2:e28320. [PMID: 24843843 PMCID: PMC4022605 DOI: 10.4161/tisb.28320] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/16/2014] [Accepted: 02/21/2014] [Indexed: 12/19/2022] Open
Abstract
Intestinal epithelial tight junctions (TJs) are a specialized structure that determines the cell polarity and prevents the diffusion of toxins, allergens, and pathogens from the lumen into the tissue. TJs are highly dynamic and its constituent protein complexes undergo continuously remodeling and turnover under tight regulation by numerous extracellular and intracellular factors. RNA-binding proteins (RBPs) and microRNAs (miRNAs) regulate gene expression at the posttranscriptional level and are involved in many aspects of cellular physiology. An increasing body of evidence indicates that RBPs including HuR and CUG-binding protein 1 and miRNAs such as miR-192 modulate the stability and translation of mRNAs encoding TJ proteins and play an important role in the control of intestinal epithelial TJ barrier function. In this mini-review article, we highlight the changes in TJ expression and intestinal epithelial TJ barrier function after activation or inactivation of RBPs and miRNAs and further analyze in some detail the mechanisms through which the stability and translation of TJ mRNAs are regulated by RBPs and miRNAs.
Collapse
Affiliation(s)
- Hong Yang
- Cell Biology Group; Department of Surgery; University of Maryland School of Medicine; Baltimore, MD USA ; Baltimore Veterans Affairs Medical Center; Baltimore, MD USA
| | - Jaladanki N Rao
- Cell Biology Group; Department of Surgery; University of Maryland School of Medicine; Baltimore, MD USA ; Baltimore Veterans Affairs Medical Center; Baltimore, MD USA
| | - Jian-Ying Wang
- Cell Biology Group; Department of Surgery; University of Maryland School of Medicine; Baltimore, MD USA ; Department of Pathology; University of Maryland School of Medicine; Baltimore, MD USA ; Baltimore Veterans Affairs Medical Center; Baltimore, MD USA
| |
Collapse
|
50
|
Song HP, Li RL, Chen X, Wang YY, Cai JZ, Liu J, Chen WW. Atractylodes macrocephala Koidz promotes intestinal epithelial restitution via the polyamine--voltage-gated K+ channel pathway. JOURNAL OF ETHNOPHARMACOLOGY 2014; 152:163-172. [PMID: 24417867 DOI: 10.1016/j.jep.2013.12.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 10/30/2013] [Accepted: 12/30/2013] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atractylodes macrocephala Koidz (AMK) has been used widely as a digestive and tonic in traditional Chinese medicine. AMK has shown noteworthy promoting effect on intestinal epithelial cell migration, which might represent a promising candidate for the treatment of intestinal mucosa injury. The aim of this study was to investigate the efficacy of AMK on intestinal mucosal restitution and the underlying mechanisms via IEC-6 cell migration model. MATERIALS AND METHODS A wounding model of IEC-6 cells was induced by a single-edge razor blade along the diameter of six-well polystyrene plates. The cells were grown in control cultures and in cultures containing spermidine (5 μmol/L, SPD, reference drug), alpha-difluoromethylornithine (2.5 mmol/L, DFMO, polyamine inhibitor), AMK (50, 100, and 200 μg/mL), DFMO plus SPD and DFMO plus AMK for 24h. The membrane potential (MP) and cytosolic free Ca(2+) concentration ([Ca(2+)]cyt) were detected by flow cytometry, and polyamines content was determined via high-performance liquid chromatography (HPLC). The expression of Kv1.1 mRNA and protein levels were assessed by RT-qPCR and Western blot analysis, respectively. Cell migration assay was carried out using the Image-Pro Plus software. All of these indexes were used to evaluate the effectiveness of AMK. RESULTS (1) Treatment with AMK caused significant increases in cellular polyamines content, membrane hyperpolarization, an elevation of [Ca(2+)]cyt and an acceleration of cell migration in IEC-6 cells, as compared to control group. (2) AMK not only reversed the inhibitory effects of DFMO on the polyamines content, MP, and [Ca(2+)]cyt but also restored IEC-6 cell migration to control levels. (3) The Kv1.1 mRNA and protein expression were significantly increased by AMK treatment in control and polyamine-deficient IEC-6 cells. CONCLUSIONS The results of our current studies revealed that treatment with AMK significantly stimulates the migration of intestinal epithelial cells through polyamine-Kv1.1 channel signaling pathway, which could promote the healing of intestinal injury. These results suggest the potential usefulness of AMK to cure intestinal disorders characterized by injury and ineffective repair of the intestinal mucosa.
Collapse
Affiliation(s)
- Hou-Pan Song
- Spleen and Stomach Institute, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou 510405, PR China
| | - Ru-Liu Li
- Spleen and Stomach Institute, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou 510405, PR China.
| | - Xu Chen
- Spleen and Stomach Institute, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou 510405, PR China
| | - Yi-Yu Wang
- Spleen and Stomach Institute, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou 510405, PR China
| | - Jia-Zhong Cai
- Spleen and Stomach Institute, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou 510405, PR China
| | - Jia Liu
- Spleen and Stomach Institute, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou 510405, PR China
| | - Wei-Wen Chen
- Spleen and Stomach Institute, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou 510405, PR China; Research Center of Medicinal Plant Resource Science and Engineering, Guangzhou University of Chinese Medicine, 232 WaiHuan East Road, Guangzhou University Town, Guangzhou 510006, PR China
| |
Collapse
|