1
|
Li C, Li X, Niu M, Xiao D, Luo Y, Wang Y, Fang ZE, Zhan X, Zhao X, Fang M, Wang J, Xiao X, Bai Z. Unveiling correlations between aristolochic acids and liver cancer: spatiotemporal heterogeneity phenomenon. Chin Med 2024; 19:132. [PMID: 39342223 PMCID: PMC11439320 DOI: 10.1186/s13020-024-01003-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024] Open
Abstract
Aristolochic acids are a class of naturally occurring compounds in Aristolochiaceae that have similar structural skeletons and chemical properties. Exposure to aristolochic acids is a risk factor for severe kidney disease and urinary system cancer. However, the carcinogenicity of aristolochic acids to the liver, which is the main site of aristolochic acid metabolism, is unclear. Although the characteristic fingerprint of aristolochic acid-induced mutations has been detected in the liver and aristolochic acids are known to be hepatotoxic, whether aristolochic acids can directly cause liver cancer is yet to be verified. This review summarizes the findings of long-term carcinogenicity studies of aristolochic acids in experimental animals. We propose that spatiotemporal heterogeneity in the carcinogenicity of these phytochemicals could explain why direct evidence of aristolochic acids causing liver cancer has never been found in adult individuals. We also summarized the reported approaches to mitigate aristolochic acid-induced hepatotoxicity to better address the associated global safety issue and provide directions and recommendations for future investigation.
Collapse
Affiliation(s)
- Chengxian Li
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- Evidence-Based Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Xinyu Li
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Ming Niu
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Dake Xiao
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Ye Luo
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Yinkang Wang
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhi-E Fang
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Xiaoyan Zhan
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- National Key Laboratory of Kidney Diseases, Beijing, 100039, China
| | - Xu Zhao
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Mingxia Fang
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Jiabo Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Xiaohe Xiao
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
- National Key Laboratory of Kidney Diseases, Beijing, 100039, China.
| | - Zhaofang Bai
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
- National Key Laboratory of Kidney Diseases, Beijing, 100039, China.
| |
Collapse
|
2
|
Zhao R, Hu Z, Zhang X, Huang S, Yu G, Wu Z, Yu W, Lu J, Ruan B. The oncogenic mechanisms of the Janus kinase-signal transducer and activator of transcription pathway in digestive tract tumors. Cell Commun Signal 2024; 22:68. [PMID: 38273295 PMCID: PMC10809652 DOI: 10.1186/s12964-023-01421-9] [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: 09/06/2023] [Accepted: 12/03/2023] [Indexed: 01/27/2024] Open
Abstract
Digestive tract tumors are heterogeneous and involve the dysregulation of multiple signaling pathways. The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway plays a notable role in the oncogenesis of digestive tract tumors. Typically activated by pro-inflammatory cytokines, it regulates important biological processes, such as cell growth, differentiation, apoptosis, immune responses, and inflammation. The aberrant activation of this pathway manifests in different forms, including mutations in JAKs, overexpression of cytokine receptors, and sustained STAT activation, and contributes to promoting the malignant characteristics of cancer cells, including uncontrolled proliferation, resistance to apoptosis, enhanced invasion and metastasis, angiogenesis, acquisition of stem-like properties, and drug resistance. Numerous studies have shown that aberrant activation of the JAK-STAT pathway is closely related to the development and progression of digestive tract tumors, contributing to tumor survival, angiogenesis, changes in the tumor microenvironment, and even immune escape processes. In addition, this signaling pathway also affects the sensitivity of digestive tract tumors to chemotherapy and targeted therapy. Therefore, it is crucial to comprehensively understand the oncogenic mechanisms underlying the JAK-STAT pathway in order to develop effective therapeutic strategies against digestive tract tumors. Currently, several JAK-STAT inhibitors are undergoing clinical and preclinical trials as potential treatments for various human diseases. However, further investigation is required to determine the role of this pathway, as well as the effectiveness and safety of its inhibitors, especially in the context of digestive tract tumors. In this review, we provide an overview of the structure, classic activation, and negative regulation of the JAK-STAT pathway. Furthermore, we discuss the pathogenic mechanisms of JAK-STAT signaling in different digestive tract tumors, with the aim of identifying potential novel therapeutic targets. Video Abstract.
Collapse
Affiliation(s)
- Ruihong Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Zhangmin Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Xiaoli Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Shujuan Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Guodong Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Zhe Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Wei Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China.
| | - Bing Ruan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
3
|
Wen Y, Lu X, Privratsky JR, Ren J, Ali S, Yang B, Rudemiller NP, Zhang J, Nedospasov SA, Crowley SD. TNF- α from the Proximal Nephron Exacerbates Aristolochic Acid Nephropathy. KIDNEY360 2024; 5:44-56. [PMID: 37986166 PMCID: PMC10833606 DOI: 10.34067/kid.0000000000000314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
Key Points Proximal tubular TNF aggravates kidney injury and fibrogenesis in aristolochic acid nephropathy. Tubular TNF disrupts the cell cycle in injured tubular epithelial cells. TNF-mediated toxic renal injury is independent of systemic immune responses. Background Aristolochic acid nephropathy (AAN) presents with tubular epithelial cell (TEC) damage and tubulointerstitial inflammation. Although TNF-α regulates cell apoptosis and inflammatory responses, the effects of tubular TNF in the progression of AAN require elucidation. Methods Floxed TNF mice on the 129/SvEv background were crossed with PEPCK-Cre mice to generate PEPCK-Cre + TNF flox/flox (TNF PTKO) mice or bred with Ksp-Cre mice to generate KSP-Cre + TNF flox/flox (TNF DNKO) mice. TNF PTKO, TNF DNKO, and wild-type controls (Cre negative littermates) were subjected to acute and chronic AAN. Results Deletion of TNF in the proximal but not distal nephron attenuated kidney injury, renal inflammation, and tubulointerstitial fibrosis after acute or chronic aristolochic acid (AA) exposure. The TNF PTKO mice did not have altered numbers of infiltrating myeloid cells in AAN kidneys. Nevertheless, kidneys from AA-treated TNF PTKO mice had reduced levels of proteins involved in regulated cell death, higher proportions of TECs in the G0/G1 phase, and reduced TEC proportions in the G2/M phase. Pifithrin-α , which restores the cell cycle, abrogated differences between the wild-type and PTKO cohorts in G2/M phase arrest of TECs and kidney fibrosis after AA exposure. Conclusions TNF from the proximal but not the distal nephron propagates kidney injury and fibrogenesis in AAN in part by inducing G2/M cell cycle arrest of TECs.
Collapse
Affiliation(s)
- Yi Wen
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaohan Lu
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Jamie R. Privratsky
- Department of Anesthesiology, Durham VA and Duke University Medical Center, Durham, North Carolina
| | - Jiafa Ren
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Saba Ali
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Bo Yang
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Nathan P. Rudemiller
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Jiandong Zhang
- Division of Cardiology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sergei A. Nedospasov
- Engelhardt Institute of Molecular Biology, Moscow, Russia
- Institute of Cell Biology and Neurobiology, Universitatsmedizin, Berlin, Germany
| | - Steven D. Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| |
Collapse
|
4
|
Song H, Sontz RA, Vance MJ, Morris JM, Sheriff S, Zhu S, Duan S, Zeng J, Koeppe E, Pandey R, Thorne CA, Stoffel EM, Merchant JL. High-fat diet plus HNF1A variant promotes polyps by activating β-catenin in early-onset colorectal cancer. JCI Insight 2023; 8:e167163. [PMID: 37219942 PMCID: PMC10371337 DOI: 10.1172/jci.insight.167163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/19/2023] [Indexed: 05/24/2023] Open
Abstract
The incidence of early-onset colorectal cancer (EO-CRC) is rising and is poorly understood. Lifestyle factors and altered genetic background possibly contribute. Here, we performed targeted exon sequencing of archived leukocyte DNA from 158 EO-CRC participants, which identified a missense mutation at p.A98V within the proximal DNA binding domain of Hepatic Nuclear Factor 1 α (HNF1AA98V, rs1800574). The HNF1AA98V exhibited reduced DNA binding. To test function, the HNF1A variant was introduced into the mouse genome by CRISPR/Cas9, and the mice were placed on either a high-fat diet (HFD) or high-sugar diet (HSD). Only 1% of the HNF1A mutant mice developed polyps on normal chow; however, 19% and 3% developed polyps on the HFD and HSD, respectively. RNA-Seq revealed an increase in metabolic, immune, lipid biogenesis genes, and Wnt/β-catenin signaling components in the HNF1A mutant relative to the WT mice. Mouse polyps and colon cancers from participants carrying the HNF1AA98V variant exhibited reduced CDX2 and elevated β-catenin proteins. We further demonstrated decreased occupancy of HNF1AA98V at the Cdx2 locus and reduced Cdx2 promoter activity compared with WT HNF1A. Collectively, our study shows that the HNF1AA98V variant plus a HFD promotes the formation of colonic polyps by activating β-catenin via decreasing Cdx2 expression.
Collapse
Affiliation(s)
- Heyu Song
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| | - Ricky A. Sontz
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| | - Matthew J. Vance
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| | - Julia M. Morris
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Sulaiman Sheriff
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| | - Songli Zhu
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Suzann Duan
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| | - Jiping Zeng
- Department of Urology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | | | - Ritu Pandey
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Curtis A. Thorne
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Elena M. Stoffel
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Juanita L. Merchant
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, and
| |
Collapse
|
5
|
An insight on microbial degradation of benzo[a]pyrene: current status and advances in research. World J Microbiol Biotechnol 2022; 38:61. [PMID: 35199223 DOI: 10.1007/s11274-022-03250-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/11/2022] [Indexed: 12/21/2022]
Abstract
Benzo[a]pyrene (BaP) is a high molecular weight polycyclic aromatic hydrocarbon produced as a result of incomplete combustion of organic substances. Over the years, the release of BaP in the atmosphere has increased rapidly, risking human lives. BaP can form bonds with DNA leading to the formation of DNA adducts thereby causing cancer. Therefore addressing the problem of its removal from the environment is quite pertinent though it calls for a very cumbersome and tedious process owing to its recalcitrant nature. To resolve such issues many efforts have been made to develop physical and chemical technologies of BaP degradation which have neither been cost-effective nor eco-friendly. Microbial degradation of BaP, on the other hand, has gained much attention due to added advantage of the high level of microbial diversity enabling great potential to degrade the substance without impairing environmental sustainability. Microorganisms produce enzymes like oxygenases, hydrolases and cytochrome P450 that enable BaP degradation. However, microbial degradation of BaP is restricted due to several factors related to its bio-availability and soil properties. Technologies like bio-augmentation and bio-stimulation have served to enhance the degradation rate of BaP. Besides, advanced technologies such as omics and nano-technology have opened new doors for a better future of microbial degradation of BaP and related compounds.
Collapse
|
6
|
Simile MM, Cigliano A, Paliogiannis P, Daino L, Manetti R, Feo CF, Calvisi DF, Feo F, Pascale RM. Nuclear localization dictates hepatocarcinogenesis suppression by glycine N-methyltransferase. Transl Oncol 2022; 15:101239. [PMID: 34649149 PMCID: PMC8517931 DOI: 10.1016/j.tranon.2021.101239] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND GNMT (glycine N-methyltransferase) is a tumor suppressor gene, but the mechanisms mediating its suppressive activity are not entirely known. METHODS We investigated the oncosuppressive mechanisms of GNMT in human hepatocellular carcinoma (HCC). GNMT mRNA and protein levels were evaluated by quantitative RT-PCR and immunoblotting. GNMT effect in HCC cell lines was modulated through GNMT cDNA induced overexpression or anti-GNMT siRNA transfection. RESULTS GNMT was expressed at low level in human HCCs with a better prognosis (HCCB) while it was almost absent in fast-growing tumors (HCCP). In HCCB, the nuclear localization of the GNMT protein was much more pronounced than in HCCP. In Huh7 and HepG2 cell lines, GNMT forced expression inhibited the proliferation and promoted apoptosis. At the molecular level, GNMT overexpression inhibited the expression of CYP1A (Cytochrome p450, aromatic compound-inducible), PREX2 (Phosphatidylinositol-3,4,5-trisphosphate-dependent Rac exchange factor 2), PARP1 [Poly (ADP-ribose) polymerase 1], and NFKB (nuclear factor-kB) genes. By chromatin immunoprecipitation, we found GNMT binding to the promoters of CYP1A1, PREX2, PARP1, and NFKB genes resulting in their strong inhibition. These genes are implicated in hepatocarcinogenesis, and are involved in the GNMT oncosuppressive action. CONCLUSION Overall, the present data indicate that GNMT exerts a multifaceted suppressive action by interacting with various cancer-related genes and inhibiting their expression.
Collapse
Affiliation(s)
- Maria M Simile
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, Italy.
| | - Antonio Cigliano
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, Italy
| | - Panagiotis Paliogiannis
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, Italy
| | - Lucia Daino
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, Italy.
| | - Roberto Manetti
- Department of Medical, Surgical and Experimental Medicine, Medical Division, Italy
| | - Claudio F Feo
- Department of Medical, Surgical and Experimental Medicine, Division of Surgery, Italy.
| | - Diego F Calvisi
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, Italy.
| | - Francesco Feo
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, Italy.
| | - Rosa M Pascale
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, Italy.
| |
Collapse
|
7
|
Carnosine alleviates diabetic nephropathy by targeting GNMT, a key enzyme mediating renal inflammation and fibrosis. Clin Sci (Lond) 2021; 134:3175-3193. [PMID: 33241846 PMCID: PMC7726623 DOI: 10.1042/cs20201207] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/22/2020] [Accepted: 11/25/2020] [Indexed: 12/29/2022]
Abstract
Diabetic nephropathy (DN) is a common microvascular complication of diabetes and the main cause of end-stage nephropathy (ESRD). Inflammation and fibrosis play key roles in the development and progression of diabetic nephropathy. By using in vivo and in vitro DN models, our laboratory has identified the protective role of carnosine (CAR) on renal tubules. Our results showed that carnosine restored the onset and clinical symptoms as well as renal tubular injury in DN. Furthermore, carnosine decreased kidney inflammation and fibrosis in DN mice. These results were consistent with high glucose (HG)-treated mice tubular epithelial cells (MTECs). Using web-prediction algorithms, cellular thermal shift assay (CETSA) and molecular docking, we identified glycine N-methyltransferase (GNMT) as a carnosine target. Importantly, we found that GNMT, a multiple functional protein that regulates the cellular pool of methyl groups by controlling the ratio of S-adenosylmethionine (SAM) to S-adenosylhomocysteine (SAH), was down-regulated significantly in the serum of Type 1 DM patients and renal tissues of DN mice. Moreover, using cultured TECs, we confirmed that the increased GNMT expression by transient transfection mimicked the protective role of carnosine in reducing inflammation and fibrosis. Conversely, the inhibition of GNMT expression abolished the protective effects of carnosine. In conclusion, carnosine might serve as a promising therapeutic agent for DN and GNMT might be a potential therapeutic target for DN.
Collapse
|
8
|
Mitoquinone alleviates vincristine-induced neuropathic pain through inhibiting oxidative stress and apoptosis via the improvement of mitochondrial dysfunction. Biomed Pharmacother 2020; 125:110003. [PMID: 32187955 DOI: 10.1016/j.biopha.2020.110003] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/16/2022] Open
Abstract
Chemotherapy drugs such as vincristine (Vin) could cause neuropathic pain. However, it is still lack of ideal therapeutic strategy to treat it. Mitochondrial dysfunction has been involved in the pathogenesis of neuropathic pain. The mitochondrial-targeted antioxidant, mitoquinone (MitoQ), is able to modify mitochondrial signaling, showing beneficial effects on various diseases. In the study, we investigated whether MitoQ could regulate Vin-induced neuropathic pain, and the underlying molecular mechanisms. The results showed that MitoQ significantly improved Vin-induced pain hypersensitivity and glial activation in mice. In addition, Vin resulted in severe oxidative stress in spinal cord tissues of mice, which were inhibited by MitoQ treatment through improving Nrf2 (NF-E2-related factor 2) expression in nuclear. Also, MitoQ treatment dose-dependently reduced the expression of pro-inflammatory cytokines, indicating its anti-inflammatory effects. Importantly, Vin stimulation contributed to mitochondrial fission, as evidenced by the increased expression of phosphorylated Drp1 (dynamin related protein 1) and Fis (mitochondrial fission protein 1), whereas mitochondrial fussion was inhibited. However, these effects were notably abrogated by MitoQ, subsequently improving mitochondrial dysfunction. Moreover, neuron death evoked by Vin was significantly rescued by MitoQ treatment. We also observed significantly reduced expression of cleaved Caspase-3 and Bax expression in spinal cord of MitoQ-treated mice with Vin stimulation. In contrast, anti-apoptotic factor Bcl-2 protein levels decreased by Vin were restored by MitoQ. The process of Cyto-c release from mitochondria triggered by Vin was effectively inhibited in mice treated with MitoQ. These in vivo results were further verified in the primary neurons using the in vitro and ex vivo experiments. Furthermore, MitoQ treatment alleviated axonal degeneration and mitochondria dysfunction induced by Vin. Thus, mitoquinone could alleviate vincristine-induced neuropathic pain by inhibiting oxidative stress and apoptosis via the improvement of mitochondrial dysfunction.
Collapse
|
9
|
Anger EE, Yu F, Li J. Aristolochic Acid-Induced Nephrotoxicity: Molecular Mechanisms and Potential Protective Approaches. Int J Mol Sci 2020; 21:E1157. [PMID: 32050524 PMCID: PMC7043226 DOI: 10.3390/ijms21031157] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/21/2020] [Accepted: 02/05/2020] [Indexed: 12/13/2022] Open
Abstract
Aristolochic acid (AA) is a generic term that describes a group of structurally related compounds found in the Aristolochiaceae plants family. These plants have been used for decades to treat various diseases. However, the consumption of products derived from plants containing AA has been associated with the development of nephropathy and carcinoma, mainly the upper urothelial carcinoma (UUC). AA has been identified as the causative agent of these pathologies. Several studies on mechanisms of action of AA nephrotoxicity have been conducted, but the comprehensive mechanisms of AA-induced nephrotoxicity and carcinogenesis have not yet fully been elucidated, and therapeutic measures are therefore limited. This review aimed to summarize the molecular mechanisms underlying AA-induced nephrotoxicity with an emphasis on its enzymatic bioactivation, and to discuss some agents and their modes of action to reduce AA nephrotoxicity. By addressing these two aspects, including mechanisms of action of AA nephrotoxicity and protective approaches against the latter, and especially by covering the whole range of these protective agents, this review provides an overview on AA nephrotoxicity. It also reports new knowledge on mechanisms of AA-mediated nephrotoxicity recently published in the literature and provides suggestions for future studies.
Collapse
Affiliation(s)
| | | | - Ji Li
- Department of Clinical Pharmacy, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (E.E.A.); (F.Y.)
| |
Collapse
|
10
|
Tumor suppressor gene glycine N-methyltransferase and its potential in liver disorders and hepatocellular carcinoma. Toxicol Appl Pharmacol 2019; 378:114607. [PMID: 31170416 DOI: 10.1016/j.taap.2019.114607] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023]
Abstract
Glycine N-methyltransferase is a protein with many functions. In addition to catalyzing the production of sarcosine in the one carbon metabolism pathway, it plays a role in the detoxification of environmental carcinogens such as benzo[a]pyrene, aflatoxin B1, and aristocholic acid. There is also increasing evidence suggesting a role of GNMT deficiency in liver carcinogenesis. In this review, we discuss the role of GNMT in the detoxification of xenobiotics and the mechanism of GNMT suppression during liver tumorigenesis. The protective role of GNMT in the liver allows GNMT to not only serve as a marker of liver disease, but also potentially be applied in the treatment of liver disorders and hepatocellular carcinoma. We describe the potential use of GNMT in gene therapy and we introduce the development of a GNMT promoter reporter assay that can be used to screen medicinal drugs and herbal libraries for natural compounds with anti-cancer properties.
Collapse
|
11
|
Niu SW, Liang PI, Lin MY, Yeh SM, Zhen YY, Chang YH, Huang PC, Hung CC, Kuo IC, Lin HYH, Kuo MC, Li WM, Huang CN, Wu WJ, Chen LT, Chiu YW, Hwang SJ. Predominant global glomerulosclerosis in patients of upper urinary tract urothelial carcinoma with pre-existing renal function impairment is a predictor of poor renal outcomes. BMC Cancer 2019; 19:337. [PMID: 30961555 PMCID: PMC6454684 DOI: 10.1186/s12885-019-5414-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 02/27/2019] [Indexed: 12/19/2022] Open
Abstract
Background Incidence of renal dysfunction and risks of progression to end-stage renal disease (ESRD) were reported higher in upper urinary tract urothelial carcinoma (UTUC) than in renal cell carcinoma (RCC) patients after unilateral nephrectomy. Methods Totally 193 renal cancer patients, including 132 UTUC and 61 RCC, were studied to clarify whether the pathological changes of the kidney remnant removed from nephrectomy and the clinical factors might predict the risk of ESRD. Renal tubulointerstitial (TI) score and global glomerulosclerosis (GGS) rate were examined by one pathologist and two nephrologists independently under same histopathological criteria. Results The glomerular filtration rates at the time of surgery were lower in UTUC than RCC groups (p < 0.001). Average GGS score and average TI rate were higher in UTUC than in RCC groups (p < 0.001; p < 0.001). Competitive risk factor analysis revealed that abnormal GGS rate not related to age, predominant in UTUC with pre-existing renal function impairment, was a histopathological predictor of poor renal outcomes (creatinine doubling or ESRD) within 5 years in UTUC patients. Conclusion Pre-existing renal function and pathological change of kidney remnant in both UTUC and RCC have the value for prediction of renal outcomes. Electronic supplementary material The online version of this article (10.1186/s12885-019-5414-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sheng-Wen Niu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan.,Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tzyou 1st Road, 80708, Kaohsiung, Taiwan.,Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Peir-In Liang
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Yen Lin
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tzyou 1st Road, 80708, Kaohsiung, Taiwan.,Master of Public Health Degree Program, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Shih-Meng Yeh
- Yozen clinic, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Yi Zhen
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tzyou 1st Road, 80708, Kaohsiung, Taiwan
| | - Yu-Han Chang
- Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pin-Chia Huang
- Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Chi Hung
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tzyou 1st Road, 80708, Kaohsiung, Taiwan.,Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-Ching Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan.,Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tzyou 1st Road, 80708, Kaohsiung, Taiwan.,Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hugo You-Hsien Lin
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tzyou 1st Road, 80708, Kaohsiung, Taiwan.,Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Chuan Kuo
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Ming Li
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Nung Huang
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Jeng Wu
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Yi-Wen Chiu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tzyou 1st Road, 80708, Kaohsiung, Taiwan.,Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shang-Jyh Hwang
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tzyou 1st Road, 80708, Kaohsiung, Taiwan. .,Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
12
|
Yang MH, Liao CC, Hung JH, Lai XT, Yen CH, Chen YMA. Utilizing proteomic approach to identify nuclear translocation related serine kinase phosphorylation site of GNMT as downstream effector for benzo[a]pyrene. J Food Drug Anal 2019; 27:603-609. [PMID: 30987732 PMCID: PMC9296196 DOI: 10.1016/j.jfda.2018.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 01/18/2023] Open
Abstract
Glycine N-methyltransferase (GNMT) protein is highly expressed in certain tissues, such as liver, pancreas, and prostate. GNMT serves multiple roles which include a methyl group transfer enzyme and a liver tumor suppressor. Benzo(a)pyrene (BaP), a family member of polycyclic aromatic hydrocarbon (PAH), is a known environmental carcinogen found in coal tar, tobacco smoke, barbecued food and incomplete combustion of auto fuel. BaP recruits cytochrome P450 to transform itself into benzo(a)pyrene-7,8-diol-9,10-epoxide (B(a) PDE), which covalently interacts with DNA causing tumorigenesis. BaP can be detoxified through GNMT and induces GNMT translocation into the cellular nucleus. GNMT translocation is accompanied by phosphorylation, but the role of phosphorylation in GNMT remains to be explored. Using liquid chromatography coupled with tandem mass spectrometry, this study identified serine 9 of GNMT as the phosphorylation site upon BaP treatment. When serine 9 was mutated and lost the capability to be phosphorylated, the occurrence of BaP-induced GNMT nuclear translocation was dramatically decreased. Also, this mutant from of GNMT lost the ability of phosphorylation and increased cytochrome P450 1A1 (Cyp1a) expression upon BaP treatment. In addition, protein kinase C (PKC) and c-Jun NH2-terminal kinase (JNK) may be required for such phosphorylation. Further characterization of phosphorylated GNMT for its link to BaP may bring new insights into chemical detoxification.
Collapse
Affiliation(s)
- Ming-Hui Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529,
Taiwan
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung 80708,
Taiwan
- Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, 11031,
Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807,
Taiwan
| | - Chen-Chung Liao
- Proteomics Research Center, National Yang-Ming University, Taipei 11221,
Taiwan
| | - Jung-Hsien Hung
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung 80708,
Taiwan
| | - Xiu-Ting Lai
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 11221,
Taiwan
| | - Chia-Hung Yen
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung 80708,
Taiwan
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708,
Taiwan
- Research Center for Natural products and Drug Development, Kaohsiung Medical University, Kaohsiung,
Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung,
Taiwan
- Corresponding author. E-mail addresses: (C.-H. Yen), (Y.-M.A. Chen)
| | - Yi-Ming Arthur Chen
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung 80708,
Taiwan
- Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, 11031,
Taiwan
- Corresponding author. No. 100, Shih-Chuan Ist Rd, Kaohsiung City, 80708, Taiwan. Fax: +886 7 3222783
| |
Collapse
|
13
|
MicroRNA-224 down-regulates Glycine N-methyltransferase gene expression in Hepatocellular Carcinoma. Sci Rep 2018; 8:12284. [PMID: 30115977 PMCID: PMC6095880 DOI: 10.1038/s41598-018-30682-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 07/06/2018] [Indexed: 12/11/2022] Open
Abstract
Glycine N-methyltransferase (GNMT) is a tumor suppressor for HCC. It is down-regulated in HCC, but the mechanism is not fully understood. MicroRNA-224 (miR-224) acts as an onco-miR in HCC. This study is the first to investigate miR-224 targeting the coding region of GNMT transcript. The GNMT-MT plasmid containing a miR-224 binding site silent mutation of the GNMT coding sequence can escape the suppression of miR-224 in HEK293T cells. Expression of both exogenous and endogenous GNMT was suppressed by miR-224, while miR-224 inhibitor enhanced GNMT expression. miR-224 counteracts the effects of GNMT on the reduction of cell proliferation and tumor growth. The levels of miR-224 and GNMT mRNA showed a significant inverse relationship in tumor specimens from HCC patients. Utilizing CCl4-treated hepatoma cells and mice as a cell damage of inflammatory or liver injury model, we observed that the decreased expression levels of GNMT were accompanied with the elevated expression levels of miR-224 in hepatoma cells and mouse liver. Finally, hepatic AAV-mediated GNMT also reduced CCl4-induced miR-224 expression and liver fibrosis. These results indicated that AAV-mediated GNMT has potential liver protection activity. miR-224 can target the GNMT mRNA coding sequence and plays an important role in GNMT suppression during liver tumorigenesis.
Collapse
|