1
|
Soumoy L, Genbauffe A, Mouchart L, Sperone A, Trelcat A, Mukeba-Harchies L, Wells M, Blankert B, Najem A, Ghanem G, Saussez S, Journe F. ATP1A1 is a promising new target for melanoma treatment and can be inhibited by its physiological ligand bufalin to restore targeted therapy efficacy. Cancer Cell Int 2024; 24:8. [PMID: 38178183 PMCID: PMC10765859 DOI: 10.1186/s12935-023-03196-y] [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: 07/30/2023] [Accepted: 12/26/2023] [Indexed: 01/06/2024] Open
Abstract
Despite advancements in treating metastatic melanoma, many patients exhibit resistance to targeted therapies. Our study focuses on ATP1A1, a sodium pump subunit associated with cancer development. We aimed to assess ATP1A1 prognostic value in melanoma patients and examine the impact of its ligand, bufalin, on melanoma cell lines in vitro and in vivo. High ATP1A1 expression (IHC) correlated with reduced overall survival in melanoma patients. Resistance to BRAF inhibitor was linked to elevated ATP1A1 levels in patient biopsies (IHC, qPCR) and cell lines (Western blot, qPCR). Additionally, high ATP1A1 mRNA expression positively correlated with differentiation/pigmentation markers based on data from The Cancer Genome Atlas (TCGA) databases and Verfaillie proliferative gene signature analysis. Bufalin specifically targeted ATP1A1 in caveolae, (proximity ligation assay) and influenced Src phosphorylation (Western blot), thereby disrupting multiple signaling pathways (phosphokinase array). In vitro, bufalin induced apoptosis in melanoma cell lines by acting on ATP1A1 (siRNA experiments) and, in vivo, significantly impeded melanoma growth using a nude mouse xenograft model with continuous bufalin delivery via an osmotic pump. In conclusion, our study demonstrates that ATP1A1 could serve as a prognostic marker for patient survival and a predictive marker for response to BRAF inhibitor therapy. By targeting ATP1A1, bufalin inhibited cell proliferation, induced apoptosis in vitro, and effectively suppressed tumor development in mice. Thus, our findings strongly support ATP1A1 as a promising therapeutic target, with bufalin as a potential agent to disrupt its tumor-promoting activity.
Collapse
Affiliation(s)
- Laura Soumoy
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium.
- Institut National de la Santé et de la Recherche Médicale (INSERM) U981, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Aline Genbauffe
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium
| | - Lena Mouchart
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium
| | - Alexandra Sperone
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium
| | - Anne Trelcat
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium
| | - Léa Mukeba-Harchies
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium
| | - Mathilde Wells
- Laboratory of Pharmaceutical Analysis, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium
| | - Bertrand Blankert
- Laboratory of Pharmaceutical Analysis, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium
| | - Ahmad Najem
- Laboratory of Clinical and Experimental Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1000, Brussels, Belgium
| | - Ghanem Ghanem
- Laboratory of Clinical and Experimental Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1000, Brussels, Belgium
| | - Sven Saussez
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium
- Department of Otolaryngology and Head and Neck Surgery, CHU Saint-Pierre, 1000, Brussels, Belgium
| | - Fabrice Journe
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium.
- Laboratory of Clinical and Experimental Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1000, Brussels, Belgium.
| |
Collapse
|
2
|
Xu Y, Xu J, Zhu W, Yan Y, Jiang X, Xie Z, Feng F, Zhang J. Bioassay-Guided Fractionation and Biological Activity of Cardenolides from Streptocaulon juventas. PLANTA MEDICA 2023; 89:1444-1456. [PMID: 37709286 DOI: 10.1055/a-2114-5371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
The discovery that Na/K-ATPase acts as a signal transducer led us to investigate the structural diversity of cardiotonic steroids and study their ligand effects. By applying Na/K-ATPase activity assay-guided fractionation, we isolated a total of 20 cardiotonic steroids from Streptocaulon juventas, including an undescribed juventasoside B (10: ) and 19 known cardiotonic steroids. Their structures have been elucidated. Using our platform of purified Na/K-ATPase and an LLC-PK1 cell model, we found that 10: , at a concentration that induces less than 10% Na/K-ATPase inhibition, can stimulate the Na/K-ATPase/Src receptor complex and selectively activate downstream pathways, ultimately altering prostate cancer cell growth. By assessing the ligand effect of the isolated cardiotonic steroids, we found that the regulation of cell viability by the isolated cardiotonic steroids was not associated with their inhibitory potencies against Na/K-ATPase activity but reflected their ligand-binding affinity to the Na/K-ATPase receptor. Based on this discovery, we identified a unique active cardiotonic steroid, digitoxigenin (1: ), and verified that it can protect LLC-PK1 cells from hypoxic injury, implicating its potential use in ischemia/reperfusion injury and inducing collagen synthesis in primary human dermal fibroblast cells, and implicating that compound 2: is the molecular basis of the wound healing activity of S. juventas.
Collapse
Affiliation(s)
- Yunhui Xu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Jian Xu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Wanfang Zhu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Yanling Yan
- Departments of Clinical & Translational Sciences, Biomedical Sciences and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Xueyang Jiang
- Department of Medicinal Chemistry, Anhui University of Chinese Medicine, Hefei, China
| | - Zijian Xie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Jie Zhang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
3
|
Rajan PK, Udoh UAS, Nakafuku Y, Pierre SV, Sanabria J. Normalization of the ATP1A1 Signalosome Rescinds Epigenetic Modifications and Induces Cell Autophagy in Hepatocellular Carcinoma. Cells 2023; 12:2367. [PMID: 37830582 PMCID: PMC10572209 DOI: 10.3390/cells12192367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/14/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. In metabolic dysfunction-associated steatohepatitis (MASH)-related HCC, cellular redox imbalance from metabolic disturbances leads to dysregulation of the α1-subunit of the Na/K-ATPase (ATP1A1) signalosome. We have recently reported that the normalization of this pathway exhibited tumor suppressor activity in MASH-HCC. We hypothesized that dysregulated signaling from the ATP1A1, mediated by cellular metabolic stress, promotes aberrant epigenetic modifications including abnormal post-translational histone modifications and dysfunctional autophagic activity, leading to HCC development and progression. Increased H3K9 acetylation (H3K9ac) and H3K9 tri-methylation (H3K9me3) were observed in human HCC cell lines, HCC-xenograft and MASH-HCC mouse models, and epigenetic changes were associated with decreased cell autophagy in HCC cell lines. Inhibition of the pro-autophagic transcription factor FoxO1 was associated with elevated protein carbonylation and decreased levels of reduced glutathione (GSH). In contrast, normalization of the ATP1A1 signaling significantly decreased H3K9ac and H3K9me3, in vitro and in vivo, with concomitant nuclear localization of FoxO1, heightening cell autophagy and cancer-cell apoptotic activities in treated HCC cell lines. Our results showed the critical role of the ATP1A1 signalosome in HCC development and progression through epigenetic modifications and impaired cell autophagy activity, highlighting the importance of the ATP1A1 pathway as a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Pradeep Kumar Rajan
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Utibe-Abasi S. Udoh
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Yuto Nakafuku
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Sandrine V. Pierre
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Juan Sanabria
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
- Department of Nutrition and Metabolomic Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH 44100, USA
| |
Collapse
|
4
|
Sodhi K, Maxwell K, Yan Y, Liu J, Chaudhry MA, Xie Z, Shapiro JI. pNaKtide Inhibits Na/K-ATPase Signaling and Attenuates Obesity. JOURNAL OF CLINICAL AND MEDICAL SCIENCES 2023; 7:1000238. [PMID: 38283397 PMCID: PMC10812088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Obesity is a growing public health crisis across the world and has been recognized as an underlying risk factor for metabolic syndrome. Growing evidence demonstrates the critical role of oxidative stress in the pathophysiological mechanisms of obesity and related metabolic dysfunction. As we have established previously that Na/K-ATPase can amplify oxidative stress signaling, we aimed to explore the effect of inhibition of this pathway on obesity phenotype using the peptide antagonist, pNaKtide. The experiments performed in murine preadipocytes showed the dose-dependent effect of pNaKtide in attenuating oxidant stress and lipid accumulation. Furthermore, these in vitro findings were confirmed in C57Bl6 mice fed a high-fat diet. Interestingly, pNaKtide could significantly reduce body weight, ameliorate systemic oxidative and inflammatory milieu and improve insulin sensitivity in obese mice. Hence the study demonstrates the therapeutic utility of pNaKtide as an inhibitor of Na/K-ATPase oxidant amplification signaling to alleviate obesity and associated comorbidities.
Collapse
Affiliation(s)
- Komal Sodhi
- Department of Medicine, Biomedical Science, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, United States of America
| | - Kyle Maxwell
- Department of Medicine, Biomedical Science, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, United States of America
| | - Yanling Yan
- Department of Medicine, Biomedical Science, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, United States of America
| | - Jiang Liu
- Department of Medicine, Biomedical Science, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, United States of America
| | - Muhammad A Chaudhry
- Department of Medicine, Biomedical Science, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, United States of America
| | - Zijian Xie
- Department of Medicine, Biomedical Science, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, United States of America
| | - Joseph I Shapiro
- Department of Medicine, Biomedical Science, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, United States of America
| |
Collapse
|
5
|
Pillai SS, Pereira DG, Zhang J, Huang W, Beg MA, Knaack DA, de Souza Goncalves B, Sahoo D, Silverstein RL, Shapiro JI, Sodhi K, Chen Y. Contribution of adipocyte Na/K-ATPase α1/CD36 signaling induced exosome secretion in response to oxidized LDL. Front Cardiovasc Med 2023; 10:1046495. [PMID: 37180782 PMCID: PMC10174328 DOI: 10.3389/fcvm.2023.1046495] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction Adipose tissue constantly secretes adipokines and extracellular vesicles including exosomes to crosstalk with distinct tissues and organs for whole-body homeostasis. However, dysfunctional adipose tissue under chronic inflammatory conditions such as obesity, atherosclerosis, and diabetes shows pro-inflammatory phenotypes accompanied by oxidative stress and abnormal secretion. Nevertheless, molecular mechanisms of how adipocytes are stimulated to secrete exosomes under those conditions remain poorly understood. Methods Mouse and human in vitro cell culture models were used for performing various cellular and molecular studies on adipocytes and macrophages. Statistical analysis was performed using Student's t-test (two-tailed, unpaired, and equal variance) for comparisons between two groups or ANOVA followed by Bonferroni's multiple comparison test for comparison among more than two groups. Results and discussion In this work, we report that CD36, a scavenger receptor for oxidized LDL, formed a signaling complex with another membrane signal transducer Na/K-ATPase in adipocytes. The atherogenic oxidized LDL induced a pro-inflammatory response in in vitro differentiated mouse and human adipocytes and also stimulated the cells to secrete more exosomes. This was largely blocked by either CD36 knockdown using siRNA or pNaKtide, a peptide inhibitor of Na/K-ATPase signaling. These results showed a critical role of the CD36/Na/K-ATPase signaling complex in oxidized LDL-induced adipocyte exosome secretion. Moreover, by co-incubation of adipocyte-derived exosomes with macrophages, we demonstrated that oxidized LDL-induced adipocyte-derived exosomes promoted pro-atherogenic phenotypes in macrophages, including CD36 upregulation, IL-6 secretion, metabolic switch to glycolysis, and mitochondrial ROS production. Altogether, we show here a novel mechanism through which adipocytes increase exosome secretion in response to oxidized LDL and that the secreted exosomes can crosstalk with macrophages, which may contribute to atherogenesis.
Collapse
Affiliation(s)
- Sneha S. Pillai
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Duane G. Pereira
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Jue Zhang
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Wenxin Huang
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Mirza Ahmar Beg
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Darcy A. Knaack
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bruno de Souza Goncalves
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Daisy Sahoo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Roy L. Silverstein
- Versiti Blood Research Institute, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Joseph I. Shapiro
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Komal Sodhi
- Department of Surgery, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Yiliang Chen
- Versiti Blood Research Institute, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
6
|
The Alpha-1 Subunit of the Na +/K +-ATPase (ATP1A1) Is a Host Factor Involved in the Attachment of Porcine Epidemic Diarrhea Virus. Int J Mol Sci 2023; 24:ijms24044000. [PMID: 36835408 PMCID: PMC9966514 DOI: 10.3390/ijms24044000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/18/2023] Open
Abstract
Porcine epidemic diarrhea (PED) is an acute and severe atrophic enteritis caused by porcine epidemic diarrhea virus (PEDV) that infects pigs and makes huge economic losses to the global swine industry. Previously, researchers have believed that porcine aminopeptidase-N (pAPN) was the primary receptor for PEDV, but it has been found that PEDV can infect pAPN knockout pigs. Currently, the functional receptor for PEDV remains unspecified. In the present study, we performed virus overlay protein binding assay (VOPBA), found that ATP1A1 was the highest scoring protein in the mass spectrometry results, and confirmed that the CT structural domain of ATP1A1 interacts with PEDV S1. First, we investigated the effect of ATP1A1 on PEDV replication. Inhibition of hosts ATP1A1 protein expression using small interfering RNA (siRNAs) significantly reduced the cells susceptibility to PEDV. The ATP1A1-specific inhibitors Ouabain (a cardiac steroid) and PST2238 (a digitalis toxin derivative), which specifically bind ATP1A1, could block the ATP1A1 protein internalization and degradation, and consequently reduce the infection rate of host cells by PEDV significantly. Additionally, as expected, overexpression of ATP1A1 notably enhanced PEDV infection. Next, we observed that PEDV infection of target cells resulted in upregulation of ATP1A1 at the mRNA and protein levels. Furthermore, we found that the host protein ATP1A1 was involved in PEDV attachment and co-localized with PEDV S1 protein in the early stage of infection. In addition, pretreatment of IPEC-J2 and Vero-E6 cells with ATP1A1 mAb significantly reduced PEDV attachment. Our observations provided a perspective on identifying key factors in PEDV infection, and may provide valuable targets for PEDV infection, PEDV functional receptor, related pathogenesis, and the development of new antiviral drugs.
Collapse
|
7
|
Zheng J, Lan P, Meng X, Kang MC, Huang X, Yan X. Na +/K +-ATPase DR region antibody ameliorated cardiac hypertrophy and fibrosis in rats with 5/6 nephrectomy. Exp Biol Med (Maywood) 2022; 247:1785-1794. [PMID: 35833534 PMCID: PMC9638958 DOI: 10.1177/15353702221108910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The enzyme Na+/K+-ATPase (NKA) is important in the heart. Reductions in NKA activity and expression have often been observed in chronic kidney disease (CKD)-related heart injury. Previously, our group found that an antibody targeting the NKA1α1 subunit's DR extracellular region (897DVEDSYGQQWTYEQR911) stimulated NKA activities and produced cardioprotective effects against ischemic injury and isoproterenol-induced cardiac remodeling. In here, we assessed whether DRm217, a specific DR antibody, exhibits cardioprotective effects in chronic renal failure models. In 5/6 nephrectomy (5/6 Nx) surgery to mimic CKD in Sprague Dawley rat, we observed that NKA activity and expression were depressed in the hearts of 5/6 Nx rats. DRm217, an NKA DR region antibody, alleviated heart hypertrophy and cardiac fibrosis under 5/6 Nx conditions. Further studies revealed that DRm217 inhibited Src activation and reduced reactive oxygen species (ROS) levels in hearts under 5/6 Nx conditions. Our findings imply that NKA could be a treatment target in CKD-related cardiac diseases. Prevention of CKD-induced myocardial injury by DRm217 provides an appealing therapeutic alternative.
Collapse
Affiliation(s)
- Jin Zheng
- Hospital of Nephrology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, Shaanxi, China
| | - Ping Lan
- Hospital of Nephrology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, Shaanxi, China
| | - Xun Meng
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, Shaanxi, China
| | - Min-Chao Kang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, Shaanxi, China
| | - Xin Huang
- Department of Cardiology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
| | - Xiaofei Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, Shaanxi, China,Xiaofei Yan.
| |
Collapse
|
8
|
Lad A, Hunyadi J, Connolly J, Breidenbach JD, Khalaf FK, Dube P, Zhang S, Kleinhenz AL, Baliu-Rodriguez D, Isailovic D, Hinds TD, Gatto-Weis C, Stanoszek LM, Blomquist TM, Malhotra D, Haller ST, Kennedy DJ. Antioxidant Therapy Significantly Attenuates Hepatotoxicity following Low Dose Exposure to Microcystin-LR in a Murine Model of Diet-Induced Non-Alcoholic Fatty Liver Disease. Antioxidants (Basel) 2022; 11:1625. [PMID: 36009344 PMCID: PMC9404967 DOI: 10.3390/antiox11081625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 12/15/2022] Open
Abstract
We have previously shown in a murine model of Non-alcoholic Fatty Liver Disease (NAFLD) that chronic, low-dose exposure to the Harmful Algal Bloom cyanotoxin microcystin-LR (MC-LR), resulted in significant hepatotoxicity including micro-vesicular lipid accumulation, impaired toxin metabolism as well as dysregulation of the key signaling pathways involved in inflammation, immune response and oxidative stress. On this background we hypothesized that augmentation of hepatic drug metabolism pathways with targeted antioxidant therapies would improve MC-LR metabolism and reduce hepatic injury in NAFLD mice exposed to MC-LR. We chose N-acetylcysteine (NAC, 40 mM), a known antioxidant that augments the glutathione detoxification pathway and a novel peptide (pNaKtide, 25 mg/kg) which is targeted to interrupting a specific Src-kinase mediated pro-oxidant amplification mechanism. Histological analysis showed significant increase in hepatic inflammation in NAFLD mice exposed to MC-LR which was attenuated on treatment with both NAC and pNaKtide (both p ≤ 0.05). Oxidative stress, as measured by 8-OHDG levels in urine and protein carbonylation in liver sections, was also significantly downregulated upon treatment with both antioxidants after MC-LR exposure. Genetic analysis of key drug transporters including Abcb1a, Phase I enzyme-Cyp3a11 and Phase II metabolic enzymes-Pkm (Pyruvate kinase, muscle), Pklr (Pyruvate kinase, liver, and red blood cell) and Gad1 (Glutamic acid decarboxylase) was significantly altered by MC-LR exposure as compared to the non-exposed control group (all p ≤ 0.05). These changes were significantly attenuated with both pNaKtide and NAC treatment. These results suggest that MC-LR metabolism and detoxification is significantly impaired in the setting of NAFLD, and that these pathways can potentially be reversed with targeted antioxidant treatment.
Collapse
Affiliation(s)
- Apurva Lad
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Jonathan Hunyadi
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Jacob Connolly
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | | | - Fatimah K. Khalaf
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
- Department of Clinical Pharmacy, University of Alkafeel, Najaf 54001, Iraq
| | - Prabhatchandra Dube
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Shungang Zhang
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Andrew L. Kleinhenz
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - David Baliu-Rodriguez
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Dragan Isailovic
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Cara Gatto-Weis
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Lauren M. Stanoszek
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Thomas M. Blomquist
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Deepak Malhotra
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Steven T. Haller
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - David J. Kennedy
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
9
|
Bonfim Neto AP, Cardoso APMM, Silva RDS, Sousa LMMDC, Giometti IC, Binelli M, Bauersachs S, Kowalewski MP, Papa PDC. An approach to uncover the relationship between 17b-estradiol and ESR1/ESR2 ratio in the regulation of canine corpus luteum. Front Vet Sci 2022; 9:885257. [PMID: 35982918 PMCID: PMC9378837 DOI: 10.3389/fvets.2022.885257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/15/2022] [Indexed: 11/28/2022] Open
Abstract
The canine corpus luteum (CL) is able to synthetise, activate and deactivate 17b-estradiol (E2) and also expresses nuclear estrogen receptors in a time-dependent manner during diestrus. Nevertheless, we are still missing a better comprehension of E2 functions in the canine CL, especially regarding the specific roles of estrogen receptor alpha (ERa) and ERb, encoded by ESR1 and 2, respectively. For that purpose, we analyzed transcriptomic data of canine non-pregnant CL collected on days 10, 20, 30, 40, 50 and 60 of diestrus and searched for differentially expressed genes (DEG) containing predicted transcription factor binding sites (TFBS) for ESR1 or ESR2. Based on biological functions of DEG presenting TFBS, expression of select transcripts and corresponding proteins was assessed. Additionally, luteal cells were collected across specific time points during diestrus and specificity of E2 responses was tested using ERa and/or ERb inhibitors. Bioinformatic analyses revealed 517 DEGs containing TFBS, from which 67 for both receptors. In general, abundance of predicted ESR1 targets was greater in the beginning, while abundance of ESR2 targets was greater in the end of diestrus. ESR1/ESR2 ratio shifted from an increasing to a decreasing pattern from day 30 to 40 post ovulation. Specific receptor inhibition suggested an ERa-mediated positive regulation of CL function at the beginning of diestrus and an ERb-mediated effect contributing to luteal regression. In conclusion, our data points toward a broad spectrum of action of E2 and its nuclear receptors, which can also act as transcription factors for other genes regulating canine CL function.
Collapse
Affiliation(s)
| | | | - Renata dos Santos Silva
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Ines Cristina Giometti
- Faculty of Veterinary Medicine, University of Western São Paulo, Presidente Prudente, Brazil
| | - Mario Binelli
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Stefan Bauersachs
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | | | - Paula de Carvalho Papa
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- *Correspondence: Paula de Carvalho Papa
| |
Collapse
|
10
|
Src Family Kinases: A Potential Therapeutic Target for Acute Kidney Injury. Biomolecules 2022; 12:biom12070984. [PMID: 35883540 PMCID: PMC9312434 DOI: 10.3390/biom12070984] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 02/04/2023] Open
Abstract
Src family kinases (SFKs) are non-receptor tyrosine kinases and play a key role in regulating signal transduction. The mechanism of SFKs in various tumors has been widely studied, and there are more and more studies on its role in the kidney. Acute kidney injury (AKI) is a disease with complex pathogenesis, including oxidative stress (OS), inflammation, endoplasmic reticulum (ER) stress, autophagy, and apoptosis. In addition, fibrosis has a significant impact on the progression of AKI to developing chronic kidney disease (CKD). The mortality rate of this disease is very high, and there is no effective treatment drug at present. In recent years, some studies have found that SFKs, especially Src, Fyn, and Lyn, are involved in the pathogenesis of AKI. In this paper, the structure, function, and role of SFKs in AKI are discussed. SFKs play a crucial role in the occurrence and development of AKI, making them promising molecular targets for the treatment of AKI.
Collapse
|
11
|
Udoh UAS, Banerjee M, Rajan PK, Sanabria JD, Smith G, Schade M, Sanabria JA, Nakafuku Y, Sodhi K, Pierre SV, Shapiro JI, Sanabria JR. Tumor-Suppressor Role of the α1-Na/K-ATPase Signalosome in NASH Related Hepatocellular Carcinoma †. Int J Mol Sci 2022; 23:ijms23137359. [PMID: 35806364 PMCID: PMC9266688 DOI: 10.3390/ijms23137359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related mortality worldwide, with an estimate of 0.84 million cases every year. In Western countries, because of the obesity epidemic, non-alcoholic steatohepatitis (NASH) has become the major cause of HCC. Intriguingly, the molecular mechanisms underlying tumorigenesis of HCC from NASH are largely unknown. We hypothesized that the growing uncoupled metabolism during NASH progression to HCC, manifested by lower cell redox status and an apoptotic ‘switch’ activity, follows a dysregulation of α1-Na/K-ATPase (NKA)/Src signalosome. Our results suggested that in NASH-related malignancy, α1-NKA signaling causes upregulation of the anti-apoptotic protein survivin and downregulation of the pro-apoptotic protein Smac/DIABLO via the activation of the PI3K → Akt pro-survival pathway with concomitant inhibition of the FoxO3 circuit, favoring cell division and primary liver carcinogenesis. Signalosome normalization using an inhibitory peptide resets apoptotic activity in malignant cells, with a significant decrease in tumor burden in vivo. Therefore, α1-NKA signalosome exercises in HCC the characteristic of a tumor suppressor, suggesting α1-NKA as a putative target for clinical therapy.
Collapse
Affiliation(s)
- Utibe-Abasi S. Udoh
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Moumita Banerjee
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Pradeep K. Rajan
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Juan D. Sanabria
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Gary Smith
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Mathew Schade
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Jacqueline A. Sanabria
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Yuto Nakafuku
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Komal Sodhi
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Joseph I. Shapiro
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Juan R. Sanabria
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
- Department of Nutrition and Metabolomic Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Correspondence: or
| |
Collapse
|
12
|
Liu J, Chaudhry M, Bai F, Chuang J, Chaudhry H, Al-Astal AEY, Nie Y, Sollars V, Sodhi K, Seligman P, Shapiro JI. Blockage of the Na-K-ATPase signaling-mediated oxidant amplification loop elongates red blood cell half-life and ameliorates uremic anemia induced by 5/6th PNx in C57BL/6 mice. Am J Physiol Renal Physiol 2022; 322:F655-F666. [PMID: 35435001 PMCID: PMC9076417 DOI: 10.1152/ajprenal.00189.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 03/21/2022] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
We have previously demonstrated that the Na-K-ATPase signaling-mediated oxidant amplification loop contributes to experimental uremic cardiomyopathy and anemia induced by 5/6th partial nephrectomy (PNx). This process can be ameliorated by systemic administration of the peptide pNaKtide, which was designed to block this oxidant amplification loop. The present study demonstrated that the PNx-induced anemia is characterized by marked decreases in red blood cell (RBC) survival as assessed by biotinylated RBC clearance and eryptosis as assessed by annexin V binding. No significant change in iron homeostasis was observed. Examination of plasma samples demonstrated that PNx induced significant increases in systemic oxidant stress as assessed by protein carbonylation, plasma erythropoietin concentration, and blood urea nitrogen. Systemic administration of pNaKtide, but not NaKtide (pNaKtide without the TAT leader sequence) and a scramble "pNaKtide" (sc-pNaKtide), led to the normalization of hematocrit, RBC survival, and plasma protein carbonylation. Administration of the three peptides had no significant effect on PNx-induced increases in plasma erythropoietin and blood urea nitrogen without notable changes in iron metabolism. These data indicate that blockage of the Na-K-ATPase signaling-mediated oxidant amplification loop ameliorates the anemia of experimental renal failure by increasing RBC survival.NEW & NOTEWORTHY The anemia of CKD is multifactorial, and the current treatment based primarily on stimulating bone marrow production of RBCs with erythropoietin or erythropoietin analogs is unsatisfactory. In a murine model of CKD that is complicated by anemia, blockade of Na-K-ATPase signaling with a specific peptide (pNaKtide) ameliorated the anemia primarily by increasing RBC survival. Should these results be confirmed in patients, this strategy may allow for novel and potentially additive strategies to treat the anemia of CKD.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Muhammad Chaudhry
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Justin Chuang
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Hibba Chaudhry
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Ala-Eddin Yassin Al-Astal
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Ying Nie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Vincent Sollars
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Komal Sodhi
- Department of Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Paul Seligman
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Joseph I Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| |
Collapse
|
13
|
Pagano E, Elias JE, Schneditz G, Saveljeva S, Holland LM, Borrelli F, Karlsen TH, Kaser A, Kaneider NC. Activation of the GPR35 pathway drives angiogenesis in the tumour microenvironment. Gut 2022; 71:509-520. [PMID: 33758004 PMCID: PMC8862021 DOI: 10.1136/gutjnl-2020-323363] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Primary sclerosing cholangitis (PSC) is in 70% of cases associated with inflammatory bowel disease. The hypermorphic T108M variant of the orphan G protein-coupled receptor GPR35 increases risk for PSC and ulcerative colitis (UC), conditions strongly predisposing for inflammation-associated liver and colon cancer. Lack of GPR35 reduces tumour numbers in mouse models of spontaneous and colitis associated cancer. The tumour microenvironment substantially determines tumour growth, and tumour-associated macrophages are crucial for neovascularisation. We aim to understand the role of the GPR35 pathway in the tumour microenvironment of spontaneous and colitis-associated colon cancers. DESIGN Mice lacking GPR35 on their macrophages underwent models of spontaneous colon cancer or colitis-associated cancer. The role of tumour-associated macrophages was then assessed in biochemical and functional assays. RESULTS Here, we show that GPR35 on macrophages is a potent amplifier of tumour growth by stimulating neoangiogenesis and tumour tissue remodelling. Deletion of Gpr35 in macrophages profoundly reduces tumour growth in inflammation-associated and spontaneous tumour models caused by mutant tumour suppressor adenomatous polyposis coli. Neoangiogenesis and matrix metalloproteinase activity is promoted by GPR35 via Na/K-ATPase-dependent ion pumping and Src activation, and is selectively inhibited by a GPR35-specific pepducin. Supernatants from human inducible-pluripotent-stem-cell derived macrophages carrying the UC and PSC risk variant stimulate tube formation by enhancing the release of angiogenic factors. CONCLUSIONS Activation of the GPR35 pathway promotes tumour growth via two separate routes, by directly augmenting proliferation in epithelial cells that express the receptor, and by coordinating macrophages' ability to create a tumour-permissive environment.
Collapse
Affiliation(s)
- Ester Pagano
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK,Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Joshua E Elias
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK,Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Georg Schneditz
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK,Norwegian PSC Research Center, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Svetlana Saveljeva
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Lorraine M Holland
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Francesca Borrelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Tom H Karlsen
- Norwegian PSC Research Center, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Arthur Kaser
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK,Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Nicole C Kaneider
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK .,Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|
14
|
Themistocleous SC, Yiallouris A, Tsioutis C, Zaravinos A, Johnson EO, Patrikios I. Clinical significance of P-class pumps in cancer. Oncol Lett 2021; 22:658. [PMID: 34386080 PMCID: PMC8298992 DOI: 10.3892/ol.2021.12919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/12/2021] [Indexed: 12/16/2022] Open
Abstract
P-class pumps are specific ion transporters involved in maintaining intracellular/extracellular ion homeostasis, gene transcription, and cell proliferation and migration in all eukaryotic cells. The present review aimed to evaluate the role of P-type pumps [Na+/K+ ATPase (NKA), H+/K+ ATPase (HKA) and Ca2+-ATPase] in cancer cells across three fronts, namely structure, function and genetic expression. It has been shown that administration of specific P-class pumps inhibitors can have different effects by: i) Altering pump function; ii) inhibiting cell proliferation; iii) inducing apoptosis; iv) modifying metabolic pathways; and v) induce sensitivity to chemotherapy and lead to antitumor effects. For example, the NKA β2 subunit can be downregulated by gemcitabine, resulting in increased apoptosis of cancer cells. The sarcoendoplasmic reticulum calcium ATPase can be inhibited by thapsigargin resulting in decreased prostate tumor volume, whereas the HKA α subunit can be affected by proton pump inhibitors in gastric cancer cell lines, inducing apoptosis. In conclusion, the present review highlighted the central role of P-class pumps and their possible use and role as anticancer cellular targets for novel therapeutic chemical agents.
Collapse
Affiliation(s)
| | - Andreas Yiallouris
- Department of Medicine, School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus
| | - Constantinos Tsioutis
- Department of Medicine, School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus
- College of Medicine, Member of Qatar University Health, Qatar University, 2713 Doha, Qatar
| | - Elizabeth O. Johnson
- Department of Medicine, School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus
| | - Ioannis Patrikios
- Department of Medicine, School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus
| |
Collapse
|
15
|
Quantitative ubiquitylomics reveals the ubiquitination regulation landscape in oral adenoid cystic carcinoma. Biosci Rep 2021; 41:229447. [PMID: 34350460 PMCID: PMC8385350 DOI: 10.1042/bsr20211532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/04/2022] Open
Abstract
Adenoid cystic carcinoma (ACC) is an extremely rare salivary gland tumor with a poor prognosis and needs attention on molecular mechanisms. Protein ubiquitination is an evolutionarily conserved post-translational modification (PTM) for substrates degradation and controls diverse cellular functions. The broad cellular function of ubiquitination network holds great promise to detect potential targets and identify respective receptors. Novel technologies are discovered for in-depth research and characterization of the precise and dynamic regulation of ubiquitylomics in multiple cellular processes during cancer initiation, progression and treatment. In the present study, 4D label-free quantitative techniques of ubiquitination proteomics were used and we identified a total of 4152 ubiquitination sites in 1993 proteins. We also performed a systematic bioinformatics analysis for differential modified proteins and peptides containing quantitative information through the comparation between oral ACC (OACC) tumor with adjacent normal tissues, as well as the identification of eight protein clusters with motif analysis. Our findings offered an important reference of potential biomarkers and effective therapeutic targets for ACC.
Collapse
|
16
|
Liu J, Tian J, Sodhi K, Shapiro JI. The Na/K-ATPase Signaling and SGLT2 Inhibitor-Mediated Cardiorenal Protection: A Crossed Road? J Membr Biol 2021; 254:513-529. [PMID: 34297135 PMCID: PMC8595165 DOI: 10.1007/s00232-021-00192-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022]
Abstract
In different large-scale clinic outcome trials, sodium (Na+)/glucose co-transporter 2 (SGLT2) inhibitors showed profound cardiac- and renal-protective effects, making them revolutionary treatments for heart failure and kidney disease. Different theories are proposed according to the emerging protective effects other than the original purpose of glucose-lowering in diabetic patients. As the ATP-dependent primary ion transporter providing the Na+ gradient to drive other Na+-dependent transporters, the possible role of the sodium–potassium adenosine triphosphatase (Na/K-ATPase) as the primary ion transporter and its signaling function is not explored.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, JCE School of Medicine, Marshall University, Huntington, WV, USA.
| | - Jiang Tian
- Department of Biomedical Sciences, JCE School of Medicine, Marshall University, Huntington, WV, USA
| | - Komal Sodhi
- Department of Surgery, JCE School of Medicine, Marshall University, Huntington, WV, USA
| | - Joseph I Shapiro
- Departments of Medicine, JCE School of Medicine, Marshall University, Huntington, WV, USA
| |
Collapse
|
17
|
Banerjee M, Li Z, Gao Y, Lai F, Huang M, Zhang Z, Cai L, Sanabria J, Gao T, Xie Z, Pierre SV. Inverse agonism at the Na/K-ATPase receptor reverses EMT in prostate cancer cells. Prostate 2021; 81:667-682. [PMID: 33956349 PMCID: PMC10071553 DOI: 10.1002/pros.24144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/22/2020] [Accepted: 02/19/2021] [Indexed: 12/14/2022]
Abstract
The surface expression of Na/K-ATPase α1 (NKA) is significantly reduced in primary prostate tumors and further decreased in bone metastatic lesions. Here, we show that the loss of cell surface expression of NKA induces epithelial-mesenchymal transition (EMT) and promotes metastatic potential and tumor growth of prostate cancer (PCa) by decreasing the expression of E-cadherin and increasing c-Myc expression via the activation of Src/FAK pathways. Mechanistically, reduced surface expression of NKA in PCa is due to increased endocytosis through the activation of NKA/Src receptor complex. Using a high-throughput NKA ligand-screening platform, we have discovered MB5 as an inverse agonist of the NKA/Src receptor complex, capable of blocking the endocytosis of NKA. MB5 treatment increased NKA expression and E-cadherin in PCa cells, which reversed EMT and consequently decreased the invasion and growth of spheroid models and tumor xenografts. Thus, we have identified a hitherto unrecognized mechanism that regulates EMT and invasiveness of PCa and demonstrated for the first time the feasibility of identifying inverse agonists of receptor NKA/Src complex and their potential utility as anticancer drugs. We, therefore, conclude that cell surface expression of α1 NKA can be targeted for the development of new therapeutics against aggressive PCa and that MB5 may serve as a prototype for drug development against EMT in metastatic PCa.
Collapse
Affiliation(s)
- Moumita Banerjee
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, USA
| | - Zhichuan Li
- Department of Physiology, Pharmacology, and Medicine, University of Toledo Health Science Campus, Toledo, Ohio, USA
| | - Yingnyu Gao
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, USA
- Institute of Edible Fungi, Shanghai Academy of Agriculture Science, Shanghai, China
| | - Fangfang Lai
- Department of Physiology, Pharmacology, and Medicine, University of Toledo Health Science Campus, Toledo, Ohio, USA
- Institute of Materia Medica, Peking Union Medical College, Beijing, China
| | - Minqi Huang
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, USA
| | - Zhongbing Zhang
- Department of Physiology, Pharmacology, and Medicine, University of Toledo Health Science Campus, Toledo, Ohio, USA
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liquan Cai
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, USA
| | - Juan Sanabria
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, USA
- Department of Surgery, Joan Edwards School of Medicine Marshall University, Huntington, West Virginia, USA
| | - Tianyan Gao
- Department of Molecular and Cellular Biochemistry, Markey Cancer Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, USA
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, USA
| |
Collapse
|
18
|
Wang J, Wang X, Gao Y, Lin Z, Chen J, Gigantelli J, Shapiro JI, Xie Z, Pierre SV. Stress Signal Regulation by Na/K-ATPase As a New Approach to Promote Physiological Revascularization in a Mouse Model of Ischemic Retinopathy. Invest Ophthalmol Vis Sci 2021; 61:9. [PMID: 33275652 PMCID: PMC7718810 DOI: 10.1167/iovs.61.14.9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose The identification of target pathways to block excessive angiogenesis while simultaneously restoring physiological vasculature is an unmet goal in the therapeutic management of ischemic retinopathies. pNaKtide, a cell-permeable peptide that we have designed by mapping the site of α1 Na/K-ATPase (NKA)/Src binding, blocks the formation of α1 NKA/Src/reactive oxygen species (ROS) amplification loops and restores physiological ROS signaling in a number of oxidative disease models. The aim of this study was to evaluate the importance of the NKA/Src/ROS amplification loop and the effect of pNaKtide in experimental ischemic retinopathy. Methods Human retinal microvascular endothelial cells (HRMECs) and retinal pigment epithelium (ARPE-19) cells were used to evaluate the effect of pNaKtide on viability, proliferation, and angiogenesis. Retinal toxicity and distribution were assessed in those cells and in the mouse. Subsequently, the role and molecular mechanism of NKA/Src in ROS stress signaling were evaluated biochemically in the retinas of mice exposed to the well-established protocol of oxygen-induced retinopathy (OIR). Finally, pNaKtide efficacy was assessed in this model. Results The results suggest a key role of α1 NKA in the regulation of ROS stress and the Nrf2 pathway in mouse OIR retinas. Inhibition of α1 NKA/Src by pNaKtide reduced pathologic ROS signaling and restored normal expression of hypoxia-inducible factor 1-α/vascular endothelial growth factor (VEGF). Unlike anti-VEGF agents, pNaKtide did promote retinal revascularization while inhibiting neovascularization and inflammation. Conclusions Targeting α1 NKA represents a novel strategy to develop therapeutics that not only inhibit neovascularization but also promote physiological revascularization in ischemic eye diseases.
Collapse
Affiliation(s)
- Jiayan Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States.,Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States.,Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Yingnyu Gao
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Zhucheng Lin
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Jing Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - James Gigantelli
- Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Joseph I Shapiro
- Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| |
Collapse
|
19
|
Lu S, Cai S, Peng X, Cheng R, Zhang Y. Integrative Transcriptomic, Proteomic and Functional Analysis Reveals ATP1B3 as a Diagnostic and Potential Therapeutic Target in Hepatocellular Carcinoma. Front Immunol 2021; 12:636614. [PMID: 33868261 PMCID: PMC8050352 DOI: 10.3389/fimmu.2021.636614] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/12/2021] [Indexed: 12/11/2022] Open
Abstract
The Na+/K+-ATPase (NKA), has been proposed as a signal transducer involving various pathobiological processes, including tumorigenesis. However, the clinical relevance of NKA in hepatocellular carcinoma (HCC) has not been well studied. This study revealed the upregulation of mRNA of ATP1A1, ATP1B1, and ATP1B3 in HCC using TCGA, ICGC, and GEO database. Subsequently, ATP1B3 was demonstrated as an independent prognostic factor of overall survival (OS) of HCC. To investigate the potential mechanisms of ATP1B3 in HCC, we analyzed the co-expression network using LinkedOmics and found that ATP1B3 co-expressed genes were associated with immune-related biological processes. Furthermore, we found that ATP1B3 was correlated immune cell infiltration and immune-related cytokines expression in HCC. The protein level of ATP1B3 was also validated as a prognostic significance and was correlated with immune infiltration in HCC using two proteomics datasets. Finally, functional analysis revealed that ATP1B3 was increased in HCC cells and tissues, silenced ATP1B3 repressed HCC cell proliferation, migration, and promoted HCC cell apoptosis and epithelial to mesenchymal transition (EMT). In conclusion, these findings proved that ATP1B3 could be an oncogene and it was demonstrated as an independent prognostic factor and correlated with immune infiltration in HCC, revealing new insights into the prognostic role and potential immune regulation of ATP1B3 in HCC progression and provide a novel possible therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Shanshan Lu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China.,The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Shenglan Cai
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaozhen Peng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Huaihua Key Laboratory of Research and Application of Novel Molecular Diagnostic Techniques, School of Public Health & Laboratory Medicine, Hunan University of Medicine, Huaihua, China.,Department of Hunan key laboratary of aging biology, Xiangya Hospital, Central South University, Changsha, China
| | - Ruochan Cheng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Yiya Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China.,Department of Hunan key laboratary of aging biology, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
Bejček J, Spiwok V, Kmoníčková E, Rimpelová S. Na +/K +-ATPase Revisited: On Its Mechanism of Action, Role in Cancer, and Activity Modulation. Molecules 2021; 26:molecules26071905. [PMID: 33800655 PMCID: PMC8061769 DOI: 10.3390/molecules26071905] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 01/08/2023] Open
Abstract
Maintenance of Na+ and K+ gradients across the cell plasma membrane is an essential process for mammalian cell survival. An enzyme responsible for this process, sodium-potassium ATPase (NKA), has been currently extensively studied as a potential anticancer target, especially in lung cancer and glioblastoma. To date, many NKA inhibitors, mainly of natural origin from the family of cardiac steroids (CSs), have been reported and extensively studied. Interestingly, upon CS binding to NKA at nontoxic doses, the role of NKA as a receptor is activated and intracellular signaling is triggered, upon which cancer cell death occurs, which lies in the expression of different NKA isoforms than in healthy cells. Two major CSs, digoxin and digitoxin, originally used for the treatment of cardiac arrhythmias, are also being tested for another indication—cancer. Such drug repositioning has a big advantage in smoother approval processes. Besides this, novel CS derivatives with improved performance are being developed and evaluated in combination therapy. This article deals with the NKA structure, mechanism of action, activity modulation, and its most important inhibitors, some of which could serve not only as a powerful tool to combat cancer, but also help to decipher the so-far poorly understood NKA regulation.
Collapse
Affiliation(s)
- Jiří Bejček
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic; (J.B.); (V.S.)
| | - Vojtěch Spiwok
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic; (J.B.); (V.S.)
| | - Eva Kmoníčková
- Department of Pharmacology, Second Faculty of Medicine, Charles University, Plzeňská 311, 150 00 Prague, Czech Republic;
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic; (J.B.); (V.S.)
- Faculty of Medicine in Pilsen, Charles University, Alej Svobody 76, 323 00 Pilsen, Czech Republic
- Correspondence: ; Tel.: +420-220-444-360
| |
Collapse
|
21
|
Agalakova NI, Kolodkin NI, Adair CD, Trashkov AP, Bagrov AY. Preeclampsia: Cardiotonic Steroids, Fibrosis, Fli1 and Hint to Carcinogenesis. Int J Mol Sci 2021; 22:ijms22041941. [PMID: 33669287 PMCID: PMC7920043 DOI: 10.3390/ijms22041941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/31/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022] Open
Abstract
Despite prophylaxis and attempts to select a therapy, the frequency of preeclampsia does not decrease and it still takes the leading position in the structure of maternal mortality and morbidity worldwide. In this review, we present a new theory of the etiology and pathogenesis of preeclampsia that is based on the interaction of Na/K-ATPase and its endogenous ligands including marinobufagenin. The signaling pathway of marinobufagenin involves an inhibition of transcriptional factor Fli1, a negative regulator of collagen synthesis, followed by the deposition of collagen in the vascular tissues and altered vascular functions. Moreover, in vitro and in vivo neutralization of marinobufagenin is associated with the restoration of Fli1. The inverse relationship between marinobufagenin and Fli1 opens new possibilities in the treatment of cancer; as Fli1 is a proto-oncogene, a hypothesis on the suppression of Fli1 by cardiotonic steroids as a potential anti-tumor therapeutic strategy is discussed as well. We propose a novel therapy of preeclampsia that is based on immunoneutralization of the marinobufagenin by monoclonal antibodies, which is capable of impairing marinobufagenin-Na/K-ATPase interactions.
Collapse
Affiliation(s)
- Natalia I. Agalakova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, 44 Torez Prospect, 194223 St. Petersburg, Russia;
| | - Nikolai I. Kolodkin
- State Institute of Highly Pure Biopreparations and Sechenov Institute of Evolutionary Physiology and Biochemistry, 44 Torez Prospect, 194223 St. Petersburg, Russia; or
| | - C. David Adair
- Department of Obstetrics and Gynecology, University of Tennessee, Chattanooga, TN 37402, USA; or
| | - Alexander P. Trashkov
- Konstantinov St. Petersburg Nuclear Physics Institute, National Research Centre Kurchatov Institute, 1 Orlova Roshcha, 188300 Gatchina, Russia;
| | - Alexei Y. Bagrov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, 44 Torez Prospect, 194223 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
22
|
Xu Y, Jiang X, Xu J, Qu W, Xie Z, Jiang RW, Feng F. A previously undescribed phenylethanoid glycoside from Callicarpa kwangtungensis Chun acts as an agonist of the Na/K-ATPase signal transduction pathway. PHYTOCHEMISTRY 2021; 181:112577. [PMID: 33190100 DOI: 10.1016/j.phytochem.2020.112577] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 06/11/2023]
Abstract
The new concept that Na/K-ATPase acts as a receptor prompted us to look for new ligands from Callicarpa kwangtungensis Chun. Using column chromatography, an undescribed phenethyl alcohol glycoside, callicarpanoside A, and an undescribed benzyl alcohol glycoside, callicarpanoside B, along with twelve known polyphenols were isolated from Callicarpa kwangtungensis Chun. All the isolated compounds were evaluated for their Na/K-ATPase (NKA) inhibitory activities. Using our NKA technology platform-based screening assay protocols, callicarpanoside B was identified as an undescribed Na/K-ATPase agonist. In particular, the newly identified benzyl alcohol glycoside was found to bind NKA and activate the receptor NKA/Src complex, resulting in the activation of protein kinase cascades. These cascades included extracellular signal-regulated kinases and protein kinase C epsilon, as well as NKA α1 endocytosis at nanomolar concentrations. Unlike the class of cardiotonic steroids, callicarpanoside B showed less inhibition of NKA activity and caused less cellular toxicity. Moreover, callicarpanoside B was found to bind NKA at a different site other than the cardiotonic steroids binding site. Thus, we have identified an undescribed NKA α1 agonist that may be used to enhance the physiological processes of NKA α1 signaling.
Collapse
Affiliation(s)
- Yunhui Xu
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, 25701, United States
| | - Xueyang Jiang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Food and Pharmaceutical Science College, Huai'an, 223003, PR China
| | - Jian Xu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Food and Pharmaceutical Science College, Huai'an, 223003, PR China
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Food and Pharmaceutical Science College, Huai'an, 223003, PR China
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, 25701, United States
| | - Ren-Wang Jiang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Food and Pharmaceutical Science College, Huai'an, 223003, PR China.
| |
Collapse
|
23
|
Maxwell KD, Chuang J, Chaudhry M, Nie Y, Bai F, Sodhi K, Liu J, Shapiro JI. The potential role of Na-K-ATPase and its signaling in the development of anemia in chronic kidney disease. Am J Physiol Renal Physiol 2020; 320:F234-F242. [PMID: 33356956 DOI: 10.1152/ajprenal.00244.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is one of the most prominent diseases affecting our population today. According to the Factsheet published by Centers for Disease Control and Prevention (CDC), it effects approximately 15% of the total population in the United States in some way, shape, or form. Within the myriad of symptomatology associated with CKD, one of the most prevalent factors in terms of affecting quality of life is anemia. Anemia of CKD cannot be completely attributed to one mechanism or cause, but rather has a multifactorial origin in the pathophysiology of CKD. While briefly summarizing well-documented risk factors, this review, as a hypothesis, aims to explore the possible role of Na-K-ATPase and its signaling function [especially recent identified reactive oxygen species (ROS) amplification function] in the interwoven mechanisms of development of the anemia of CKD.
Collapse
Affiliation(s)
- Kyle D Maxwell
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Justin Chuang
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Muhammad Chaudhry
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Ying Nie
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Fang Bai
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Komal Sodhi
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia.,Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Jiang Liu
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Joseph I Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| |
Collapse
|
24
|
Abstract
The delivery of therapies to the cochlea is notoriously challenging. It is an organ protected by a number of barriers that need to be overcome in the drug delivery process. Additionally, there are multiple sites of possible damage within the cochlea. Despite the many potential sites of damage, acquired otologic insults preferentially damage a single location. While progress has been made in techniques for inner ear drug delivery, the current techniques remain non-specific and our ability to deliver therapies in a cell-specific manner are limited. Fortunately, there are proteins specific to various cell-types within the cochlea (e.g., hair cells, spiral ganglion cells, stria vascularis) that function as biomarkers of site-specific damage. These protein biomarkers have potential to serve as targets for cell-specific inner ear drug delivery. In this manuscript, we review the concept of biomarkers and targeted- inner ear drug delivery and the well-characterized protein biomarkers within each of the locations of interest within the cochlea. Our review will focus on targeted drug delivery in the setting of acquired otologic insults (e.g., ototoxicity, noise-induce hearing loss). The goal is not to discuss therapies to treat acquired otologic insults, rather, to establish potential concepts of how to deliver therapies in a targeted, cell-specific manner. Based on our review, it is clear that future of inner ear drug delivery is a discipline filled with potential that will require collaborative efforts among clinicians and scientists to optimize treatment of otologic insults. Graphical Abstract ![]()
Collapse
|
25
|
Wang X, Cai L, Xie JX, Cui X, Zhang J, Wang J, Chen Y, Larre I, Shapiro JI, Pierre SV, Wu D, Zhu GZ, Xie Z. A caveolin binding motif in Na/K-ATPase is required for stem cell differentiation and organogenesis in mammals and C. elegans. SCIENCE ADVANCES 2020; 6:eaaw5851. [PMID: 32537485 PMCID: PMC7253156 DOI: 10.1126/sciadv.aaw5851] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/02/2020] [Indexed: 05/15/2023]
Abstract
Several signaling events have been recognized as essential for regulating cell lineage specification and organogenesis in animals. We find that the gain of an amino-terminal caveolin binding motif (CBM) in the α subunit of the Na/K-adenosine triphosphatase (ATPase) (NKA) is required for the early stages of organogenesis in both mice and Caenorhabditis elegans. The evolutionary gain of the CBM occurred at the same time as the acquisition of the binding sites for Na+/K+. Loss of this CBM does not affect cell lineage specification or the initiation of organogenesis, but arrests further organ development. Mechanistically, this CBM is essential for the dynamic operation of Wnt and the timely up-regulation of transcriptional factors during organogenesis. These results indicate that the NKA was evolved as a dual functional protein that works in concert with Wnt as a hitherto unrecognized common mechanism to enable stem cell differentiation and organogenesis in multicellular organisms within the animal kingdom.
Collapse
Affiliation(s)
- Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research (MIIR) at Marshall University, Huntington, WV, USA
| | - Liquan Cai
- Marshall Institute for Interdisciplinary Research (MIIR) at Marshall University, Huntington, WV, USA
| | - Jeffrey X. Xie
- University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research (MIIR) at Marshall University, Huntington, WV, USA
| | - Jue Zhang
- Marshall Institute for Interdisciplinary Research (MIIR) at Marshall University, Huntington, WV, USA
| | - Jiayan Wang
- Marshall Institute for Interdisciplinary Research (MIIR) at Marshall University, Huntington, WV, USA
| | - Yiliang Chen
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, USA
| | - Isabel Larre
- Marshall Institute for Interdisciplinary Research (MIIR) at Marshall University, Huntington, WV, USA
| | - Joseph I. Shapiro
- Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research (MIIR) at Marshall University, Huntington, WV, USA
| | - Dianqing Wu
- Department of Pharmacology and Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, CT, USA
| | - Guo-Zhang Zhu
- Marshall Institute for Interdisciplinary Research (MIIR) at Marshall University, Huntington, WV, USA
- Department of Biological Sciences, Marshall University, Huntington, WV, USA
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research (MIIR) at Marshall University, Huntington, WV, USA
| |
Collapse
|
26
|
Leu WJ, Wang CT, Hsu JL, Chen IS, Chang HS, Guh JH. Ascleposide, a natural cardenolide, induces anticancer signaling in human castration-resistant prostatic cancer through Na + /K + -ATPase internalization and tubulin acetylation. Prostate 2020; 80:305-318. [PMID: 31905252 DOI: 10.1002/pros.23944] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Cardiac glycosides, which inhibit Na+ /K+ -ATPase, display inotropic effects for the treatment of congestive heart failure and cardiac arrhythmia. Recent studies have suggested signaling downstream of Na+ /K+ -ATPase action in the regulation of cell proliferation and apoptosis and have revealed the anticancer activity of cardiac glycosides. The study aims to characterize the anticancer potential of ascleposide, a natural cardenolide, and to uncover its primary target and underlying mechanism against human castration-resistant prostate cancer (CRPC). METHODS Cell proliferation was examined in CRPC PC-3 and DU-145 cells using sulforhodamine B assay, carboxyfluorescein succinimidyl ester staining assay and clonogenic examination. Flow cytometric analysis was used to detect the distribution of cell cycle phase, mitochondrial membrane potential, intracellular Na+ and Ca2+ levels, and reactive oxygen species production. Protein expression was examined using Western blot analysis. Endocytosis of Na+ /K+ -ATPase was determined using confocal immunofluorescence microscopic examination. RESULTS Ascleposide induced an increase of intracellular Na+ and a potent antiproliferative effect. It also induced a decrease of G1 phase distribution while an increase in both G2/M and apoptotic sub-G1 phases, and downregulated several cell cycle regulator proteins, including cyclins, Cdk, p21, and p27 Cip/Kip proteins, Rb and c-Myc. Ascleposide decreased the expression of antiapoptotic Bcl-2 members (eg, Bcl-2 and Mcl-1) but upregulated proapoptotic member (eg, Bak), leading to a significant loss of mitochondrial membrane potential and activation of both caspase-9 and caspase-3. Ascleposide also dramatically induced tubulin acetylation, leading to inhibition of the catalytic activity of Na+ /K+ -ATPase. Notably, extracellular high K+ (16 mM) significantly blunted ascleposide-mediated effects. Furthermore, ascleposide induced a p38 MAPK-dependent endocytosis of Na+ /K+ -ATPase and downregulated the protein expression of Na+ /K+ -ATPase α1 subunit. CONCLUSION Ascleposide displays antiproliferative and apoptotic activities dependent on the inhibition of Na+ /K+ -ATPase pumping activity through p38 MAPK-mediated endocytosis of Na+ /K+ -ATPase and downregulation of α1 subunit, which in turn cause tubulin acetylation and cell cycle arrest. Cell apoptosis is ultimately triggered by the activation of caspase cascade attributed to mitochondrial damage through the downregulation of Bcl-2 and Mcl-1 protein expressions while upregulation of Bak protein levels. The data also suggest the potential of ascleposide in anti-CRPC development.
Collapse
Affiliation(s)
- Wohn-Jenn Leu
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Ching-Ting Wang
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Jui-Ling Hsu
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Ih-Sheng Chen
- School of Pharmacy, College of Pharmacy, Kaohsiung, Taiwan, Kaohsiung, Taiwan
| | - Hsun-Shuo Chang
- School of Pharmacy, College of Pharmacy, Kaohsiung, Taiwan, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
27
|
Liu J, Nie Y, Chaudhry M, Bai F, Chuang J, Sodhi K, Shapiro JI. The Redox-Sensitive Na/K-ATPase Signaling in Uremic Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21041256. [PMID: 32069992 PMCID: PMC7072896 DOI: 10.3390/ijms21041256] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
In recent years, Na/K-ATPase signaling has been implicated in different physiological and pathophysiological conditions, including cardiac hypertrophy and uremic cardiomyopathy. Cardiotonic steroids (CTS), specific ligands of Na/K-ATPase, regulate its enzymatic activity (at higher concentrations) and signaling function (at lower concentrations without significantly affecting its enzymatic activity) and increase reactive oxygen species (ROS) generation. On the other hand, an increase in ROS alone also regulates the Na/K-ATPase enzymatic activity and signaling function. We termed this phenomenon the Na/K-ATPase-mediated oxidant-amplification loop, in which oxidative stress regulates both the Na/K-ATPase activity and signaling. Most recently, we also demonstrated that this amplification loop is involved in the development of uremic cardiomyopathy. This review aims to evaluate the redox-sensitive Na/K-ATPase-mediated oxidant amplification loop and uremic cardiomyopathy.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
- Correspondence:
| | - Ying Nie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Muhammad Chaudhry
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Justin Chuang
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| | - Komal Sodhi
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| | - Joseph I. Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| |
Collapse
|
28
|
Khalaf FK, Tassavvor I, Mohamed A, Chen Y, Malhotra D, Xie Z, Tian J, Haller ST, Westfall K, Tang WHW, Kennedy DJ. Epithelial and Endothelial Adhesion of Immune Cells Is Enhanced by Cardiotonic Steroid Signaling Through Na +/K +-ATPase-α-1. J Am Heart Assoc 2020; 9:e013933. [PMID: 32013704 PMCID: PMC7033897 DOI: 10.1161/jaha.119.013933] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Recent studies have highlighted a critical role for a group of natriuretic hormones, cardiotonic steroid (CTS), in mediating renal inflammation and fibrosis associated with volume expanded settings, such as chronic kidney disease. Immune cell adhesion is a critical step in the inflammatory response; however, little is currently understood about the potential regulatory role of CTS signaling in this setting. Herein, we tested the hypothesis that CTS signaling through Na+/K+‐ATPase α‐1 (NKA α‐1) enhances immune cell recruitment and adhesion to renal epithelium that ultimately advance renal inflammation. Methods and Results We demonstrate that knockdown of the α‐1 isoform of Na/K‐ATPase causes a reduction in CTS‐induced macrophage infiltration in renal tissue as well reduces the accumulation of immune cells in the peritoneal cavity in vivo. Next, using functional adhesion assay, we demonstrate that CTS‐induced increases in the adhesion of macrophages to renal epithelial cells were significantly diminished after reduction of NKA α‐1 in either macrophages or renal epithelial cells as well after inhibition of NKA α‐1‐Src signaling cascade with a specific peptide inhibitor, pNaKtide in vitro. Finally, CTS‐induced expression of adhesion markers in both endothelial and immune cells was significantly inhibited in an NKA α‐1‐Src signaling dependent manner in vitro. Conclusions These findings suggest that CTS potentiates immune cell migration and adhesion to renal epithelium through an NKA α‐1–dependent mechanism; our new findings suggest that pharmacological inhibition of this feed‐forward loop may be useful in the treatment of renal inflammation associated with renal disease.
Collapse
Affiliation(s)
- Fatimah K Khalaf
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Iman Tassavvor
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Amal Mohamed
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Yiliang Chen
- Blood Research Institute Blood Center of Wisconsin Milwaukee WI
| | - Deepak Malhotra
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research Marshall University Huntington WV
| | - Jiang Tian
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Steven T Haller
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Kristen Westfall
- Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Cleveland Clinic Cleveland OH
| | - W H Wilson Tang
- Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Cleveland Clinic Cleveland OH.,Center for Cardiovascular Diagnostics and Prevention Lerner Research Institute Cleveland Clinic Cleveland OH.,Department of Cardiovascular Medicine, Heart and Vascular Institute Cleveland Clinic Cleveland OH
| | - David J Kennedy
- University of Toledo College of Medicine and Life Sciences Toledo OH
| |
Collapse
|
29
|
Staehr C, Hangaard L, Bouzinova EV, Kim S, Rajanathan R, Boegh Jessen P, Luque N, Xie Z, Lykke-Hartmann K, Sandow SL, Aalkjaer C, Matchkov VV. Smooth muscle Ca 2+ sensitization causes hypercontractility of middle cerebral arteries in mice bearing the familial hemiplegic migraine type 2 associated mutation. J Cereb Blood Flow Metab 2019; 39. [PMID: 29513112 PMCID: PMC6681533 DOI: 10.1177/0271678x18761712] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Familial hemiplegic migraine type 2 (FHM2) is associated with inherited point-mutations in the Na,K-ATPase α2 isoform, including G301R mutation. We hypothesized that this mutation affects specific aspects of vascular function, and thus compared cerebral and systemic arteries from heterozygote mice bearing the G301R mutation (Atp1a2+/-G301R) with wild type (WT). Middle cerebral (MCA) and mesenteric small artery (MSA) function was compared in an isometric myograph. Cerebral blood flow was assessed with Laser speckle analysis. Intracellular Ca2+ and membrane potential were measured simultaneously. Protein expression was semi-quantified by immunohistochemistry. Protein phosphorylation was analysed by Western blot. MSA from Atp1a2+/-G301R and WT showed similar contractile responses. The Atp1a2+/-G301R MCA constricted stronger to U46619, endothelin and potassium compared to WT. This was associated with an increased depolarization, although the Ca2+ change was smaller than in WT. The enhanced constriction of Atp1a2+/-G301R MCA was associated with increased cSrc activation, stronger sensitization to [Ca2+]i and increased MYPT1 phosphorylation. These differences were abolished by cSrc inhibition. Atp1a2+/-G301R mice had reduced resting blood flow through MCA in comparison with WT mice. FHM2-associated mutation leads to elevated contractility of MCA due to sensitization of the contractile machinery to Ca2+, which is mediated via Na,K-ATPase/Src-kinase/MYPT1 signalling.
Collapse
Affiliation(s)
| | - Lise Hangaard
- 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Sukhan Kim
- 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Nathan Luque
- 2 Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Queensland, Australia
| | - Zijian Xie
- 3 Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, USA
| | | | - Shaun L Sandow
- 2 Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Queensland, Australia
| | | | | |
Collapse
|
30
|
Khalaf FK, Dube P, Kleinhenz AL, Malhotra D, Gohara A, Drummond CA, Tian J, Haller ST, Xie Z, Kennedy DJ. Proinflammatory Effects of Cardiotonic Steroids Mediated by NKA α-1 (Na+/K+-ATPase α-1)/Src Complex in Renal Epithelial Cells and Immune Cells. Hypertension 2019; 74:73-82. [PMID: 31132948 DOI: 10.1161/hypertensionaha.118.12605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiotonic steroids (CTSs) are NKA α-1 (Na+/K+-ATPase α-1) ligands that are increased in volume expanded states and associated with cardiac and renal diseases. Although initiation and resolution of inflammation is an important component of cellular injury and repair in renal disease, it is unknown whether CTS activation of NKA α-1 signaling in this setting regulates this inflammatory response. On this background, we hypothesized that CTS signaling through the NKA α-1-Src kinase complex promotes a proinflammatory response in renal epithelial and immune cells. First, we observed that the CTS telocinobufagin activated multiple proinflammatory cytokines/chemokines in renal epithelial cells, and these effects were attenuated after either NKA α-1 knockdown or with a specific inhibitor of the NKA α-1-Src kinase complex (pNaKtide). Similar findings were observed in immune cells, where we demonstrated that while telocinobufagin induced both oxidative burst and enhanced Nuclear factor kappa-light-chain-enhancer of activated B cells activation in macrophages ( P<0.05), the effects were abolished in NKA α-1+/- macrophages or by pretreatment with pNaKtide or the Src inhibitor PP2 ( P<0.01). In a series of in vivo studies, we found that 5/6th partial nephrectomy induced significantly less oxidative stress in the remnant kidney of NKA α-1+/- versus wild-type mice. Similarly, 5/6th partial nephrectomy yielded decreased levels of the urinary oxidative stress marker 8-Oxo-2'-deoxyguanosine in NKA α-1+/- versus wild-type mice. Finally, we found that in vivo inhibition of the NKA α-1-Src kinase complex with pNaKtide significantly inhibited renal proinflammatory gene expression after 5/6th partial nephrectomy. These findings suggest that the NKA α-1-Src kinase complex plays a central role in regulating the renal inflammatory response induced by elevated CTS both in vitro and in vivo.
Collapse
Affiliation(s)
- Fatimah K Khalaf
- From the Department of Medicine (F.K.K., P.D., A.L.K., D.M., C.A.D., J.T., S.T.H., D.J.K.) University of Toledo College of Medicine and Life Sciences, OH
| | - Prabhatchandra Dube
- From the Department of Medicine (F.K.K., P.D., A.L.K., D.M., C.A.D., J.T., S.T.H., D.J.K.) University of Toledo College of Medicine and Life Sciences, OH
| | - Andrew L Kleinhenz
- From the Department of Medicine (F.K.K., P.D., A.L.K., D.M., C.A.D., J.T., S.T.H., D.J.K.) University of Toledo College of Medicine and Life Sciences, OH
| | - Deepak Malhotra
- From the Department of Medicine (F.K.K., P.D., A.L.K., D.M., C.A.D., J.T., S.T.H., D.J.K.) University of Toledo College of Medicine and Life Sciences, OH
| | - Amira Gohara
- Department of Pathology (A.G.) University of Toledo College of Medicine and Life Sciences, OH
| | - Christopher A Drummond
- From the Department of Medicine (F.K.K., P.D., A.L.K., D.M., C.A.D., J.T., S.T.H., D.J.K.) University of Toledo College of Medicine and Life Sciences, OH
| | - Jiang Tian
- From the Department of Medicine (F.K.K., P.D., A.L.K., D.M., C.A.D., J.T., S.T.H., D.J.K.) University of Toledo College of Medicine and Life Sciences, OH
| | - Steven T Haller
- From the Department of Medicine (F.K.K., P.D., A.L.K., D.M., C.A.D., J.T., S.T.H., D.J.K.) University of Toledo College of Medicine and Life Sciences, OH
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV (Z.X.)
| | - David J Kennedy
- From the Department of Medicine (F.K.K., P.D., A.L.K., D.M., C.A.D., J.T., S.T.H., D.J.K.) University of Toledo College of Medicine and Life Sciences, OH
| |
Collapse
|
31
|
Abstract
The Na,K-ATPase is an enzyme essential for ion homeostasis in all cells. Over the last decades, it has been well-established that in addition to the transport of Na+/K+ over the cell membrane, the Na,K-ATPase acts as a receptor transducing humoral signals intracellularly. It has been suggested that ouabain-like compounds serve as endogenous modulators of this Na,K-ATPase signal transduction. The molecular mechanisms underlying Na,K-ATPase signaling are complicated and suggest the confluence of divergent biological pathways. This review discusses recent updates on the Na,K-ATPase signaling pathways characterized or suggested in vascular smooth muscle cells. The conventional view on this signaling is based on a microdomain structure where the Na,K-ATPase controls the Na,Ca-exchanger activity via modulation of intracellular Na+ in the spatially restricted submembrane space. This, in turn, affects intracellular Ca2+ and Ca2+ load in the sarcoplasmic reticulum leading to modulation of contractility as well as gene expression. An ion-transport-independent signal transduction from the Na,K-ATPase is based on molecular interactions. This was primarily characterized in other cell types but recently also demonstrated in vascular smooth muscles. The downstream signaling from the Na,K-ATPase includes Src and phosphatidylinositol-4,5-bisphosphate 3 kinase signaling pathways and generation of reactive oxygen species. Moreover, in vascular smooth muscle cells the interaction between the Na,K-ATPase and proteins responsible for Ca2+ homeostasis, e.g., phospholipase C and inositol triphosphate receptors, contributes to an integration of the signaling pathways. Recent update on the Na,K-ATPase dependent intracellular signaling and the significance for physiological functions and pathophysiological changes are discussed in this review.
Collapse
|
32
|
Lan YL, Zou YJ, Lou JC, Xing JS, Wang X, Zou S, Ma BB, Ding Y, Zhang B. The sodium pump α1 subunit regulates bufalin sensitivity of human glioblastoma cells through the p53 signaling pathway. Cell Biol Toxicol 2019; 35:521-539. [DOI: 10.1007/s10565-019-09462-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/18/2019] [Indexed: 12/14/2022]
|
33
|
Attenuation of Na/K-ATPase/Src/ROS amplification signal pathway with pNaktide ameliorates myocardial ischemia-reperfusion injury. Int J Biol Macromol 2018; 118:1142-1148. [DOI: 10.1016/j.ijbiomac.2018.07.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 11/21/2022]
|
34
|
Bouzinova EV, Hangaard L, Staehr C, Mazur A, Ferreira A, Chibalin AV, Sandow SL, Xie Z, Aalkjaer C, Matchkov VV. The α2 isoform Na,K-ATPase modulates contraction of rat mesenteric small artery via cSrc-dependent Ca 2+ sensitization. Acta Physiol (Oxf) 2018; 224:e13059. [PMID: 29480968 DOI: 10.1111/apha.13059] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 02/06/2018] [Accepted: 02/16/2018] [Indexed: 12/11/2022]
Abstract
AIMS The Na,K-ATPase is involved in a large number of regulatory activities including cSrc-dependent signalling. Upon inhibition of the Na,K-ATPase with ouabain, cSrc activation is shown to occur in many cell types. This study tests the hypothesis that acute potentiation of agonist-induced contraction by ouabain is mediated through Na,K-ATPase-cSrc signalling-dependent sensitization of vascular smooth muscle cells to Ca2+ . METHODS Agonist-induced rat mesenteric small artery contraction was examined in vitro under isometric conditions and in vivo in anaesthetized rats. Arterial wall tension and [Ca2+ ]i in vascular smooth muscle cells were measured simultaneously. Changes in cSrc and myosin phosphatase targeting protein 1 (MYPT1) phosphorylation were analysed by Western blot. Protein expression was examined with immunohistochemistry. The α1 and α2 isoforms of the Na,K-ATPase were transiently downregulated by siRNA transfection in vivo. RESULTS Ten micromolar ouabain, but not digoxin, potentiated contraction to noradrenaline. This effect was not endothelium-dependent. Ouabain sensitized smooth muscle cells to Ca2+ , and this was associated with increased phosphorylation of cSrc and MYPT1. Inhibition of tyrosine kinase by genistein, PP2 or pNaKtide abolished the potentiating effect of ouabain on arterial contraction and Ca2+ sensitization. Downregulation of the Na,K-ATPase α2 isoform made arterial contraction insensitive to ouabain and tyrosine kinase inhibition. CONCLUSION Data suggest that micromolar ouabain potentiates agonist-induced contraction of rat mesenteric small artery via Na,K-ATPase-dependent cSrc activation, which increases Ca2+ sensitization of vascular smooth muscle cells by MYPT1 phosphorylation. This mechanism may be critical for acute control of vascular tone.
Collapse
Affiliation(s)
- E. V. Bouzinova
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - L. Hangaard
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - C. Staehr
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - A. Mazur
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - A. Ferreira
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - A. V. Chibalin
- Department of Molecular Medicine and Surgery; Integrative Physiology; Karolinska Institutet; Stockholm Sweden
| | - S. L. Sandow
- Faculty of Science, Health, Education and Engineering; University of the Sunshine Coast; Maroochydore Qld Australia
| | - Z. Xie
- Marshall Institute for Interdisciplinary Research; Marshall University; Huntington WV USA
| | - C. Aalkjaer
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - V. V. Matchkov
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| |
Collapse
|
35
|
The Na/K-ATPase Signaling: From Specific Ligands to General Reactive Oxygen Species. Int J Mol Sci 2018; 19:ijms19092600. [PMID: 30200500 PMCID: PMC6163532 DOI: 10.3390/ijms19092600] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022] Open
Abstract
The signaling function of the Na/K-ATPase has been established for 20 years and is widely accepted in the field, with many excellent reports and reviews not cited here. Even though there is debate about the underlying mechanism, the signaling function is unquestioned. This short review looks back at the evolution of Na/K-ATPase signaling, from stimulation by cardiotonic steroids (also known as digitalis-like substances) as specific ligands to stimulation by reactive oxygen species (ROS) in general. The interplay of cardiotonic steroids and ROS in Na/K-ATPase signaling forms a positive-feedback oxidant amplification loop that has been implicated in some pathophysiological conditions.
Collapse
|
36
|
Lan YL, Yu ZL, Lou JC, Ma XC, Zhang B. Update on the effects of the sodium pump α1 subunit on human glioblastoma: from the laboratory to the clinic. Expert Opin Investig Drugs 2018; 27:753-763. [PMID: 30130132 DOI: 10.1080/13543784.2018.1512582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Glioblastoma is a debilitating disease that is associated with poor prognosis and a very limited response to therapies; thus, molecularly targeted therapeutics and personalized therapy are urgently needed. The Na+/K+-ATPase sodium pump is a transmembrane protein complex that has recently been recognized as an important transducer and integrator of various signals. The sodium pump α1 subunit, which is highly expressed in most glioblastomas compared with that in normal brain tissues, is an emerging cancer target that merits further investigation. AREAS COVERED The purpose of this narrative review is to explore the important roles of the sodium pump α1 subunit in glioblastoma and analyze its potential therapeutic applications. EXPERT OPINION Expression of the sodium pump α1 subunit in glioblastoma tissues is generally higher than that in normal tissues. Sodium pump α1 subunit-mediated pivotal antiglioblastoma signaling pathways have been reviewed, and their impact on the sensitivity of glioblastoma cells to anticancer drugs has recently been clarified. In addition, various pharmacologically optimized sodium pump inhibitors have recently reached early clinical trials, and explorations of sodium pump α1 subunit inhibitors may hold promise for the development of stratification strategies in which patients are treated based on their isoform expression status.
Collapse
Affiliation(s)
- Yu-Long Lan
- a Department of Neurosurgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , China.,b Department of Pharmacy , Dalian Medical University , Dalian , China.,c Department of Physiology , Dalian Medical University , Dalian , China
| | - Zhen-Long Yu
- b Department of Pharmacy , Dalian Medical University , Dalian , China
| | - Jia-Cheng Lou
- a Department of Neurosurgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , China
| | - Xiao-Chi Ma
- b Department of Pharmacy , Dalian Medical University , Dalian , China
| | - Bo Zhang
- a Department of Neurosurgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , China
| |
Collapse
|
37
|
Banerjee M, Cui X, Li Z, Yu H, Cai L, Jia X, He D, Wang C, Gao T, Xie Z. Na/K-ATPase Y260 Phosphorylation-mediated Src Regulation in Control of Aerobic Glycolysis and Tumor Growth. Sci Rep 2018; 8:12322. [PMID: 30120256 PMCID: PMC6098021 DOI: 10.1038/s41598-018-29995-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/23/2018] [Indexed: 12/22/2022] Open
Abstract
We report here the identification of α1 Na/K-ATPase as a major regulator of the proto-oncogene Src kinase and the role of this regulation in control of Warburg effect and tumor growth. Specifically, we discovered Y260 in α1 Na/K-ATPase as a Src-specific phosphorylation and binding site and that Y260 phosphorylation is required for Src-mediated signal transduction in response to a number of stimuli including EGF. As such, it enables a dynamic control of aerobic glycolysis. However, such regulation appears to be lost or attenuated in human cancers as the expression of Na/K-ATPase α1 was significantly decreased in prostate, breast and kidney cancers, and further reduced in corresponding metastatic lesions in patient samples. Consistently, knockdown of α1 Na/K-ATPase led to a further increase in lactate production and the growth of tumor xenograft. These findings suggest that α1 Na/K-ATPase works as a tumor suppressor and that a loss of Na/K-ATPase-mediated Src regulation may lead to Warburg phenotype in cancer.
Collapse
Affiliation(s)
- Moumita Banerjee
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA
| | - Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA
| | - Zhichuan Li
- Department of Physiology and Pharmacology and Medicine, University of Toledo College of Medicine, Toledo, Ohio, 43614, USA
| | - Hui Yu
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Liquan Cai
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA
| | - Xuelian Jia
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA
| | - Daheng He
- Department of Cancer Biostatistics, Markey Cancer Research Center, University of Kentucky, Lexington, Kentucky, 40536, USA
| | - Chi Wang
- Department of Cancer Biostatistics, Markey Cancer Research Center, University of Kentucky, Lexington, Kentucky, 40536, USA
| | - Tianyan Gao
- Department of Molecular and Cellular Biochemistry, Markey Cancer Research Center, University of Kentucky, Lexington, Kentucky, 40536, USA
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA.
| |
Collapse
|
38
|
Quantum Modeling: A Bridge between the Pumping and Signaling Functions of Na/K-ATPase. Int J Mol Sci 2018; 19:ijms19082347. [PMID: 30096926 PMCID: PMC6121303 DOI: 10.3390/ijms19082347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 12/15/2022] Open
Abstract
Although the signaling function of Na/K-ATPase has been studied for decades, the chasm between the pumping function and the signaling function of Na/K-ATPase is still an open issue. This article explores the relationship between ion pumping and signaling with attention to the amplification of oxidants through this signaling function. We specifically consider the Na/K-ATPase with respect to its signaling function as a superposition of different states described for its pumping function. We then examine how alterations in the relative amounts of these states could alter signaling through the Src-EGFR-ROS pathway. Using assumptions based on some experimental observations published by our laboratories and others, we develop some predictions regarding cellular oxidant stress.
Collapse
|
39
|
The Role of Na/K-ATPase Signaling in Oxidative Stress Related to Aging: Implications in Obesity and Cardiovascular Disease. Int J Mol Sci 2018; 19:ijms19072139. [PMID: 30041449 PMCID: PMC6073138 DOI: 10.3390/ijms19072139] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 07/19/2018] [Accepted: 07/21/2018] [Indexed: 12/12/2022] Open
Abstract
Aging has been associated with a series of pathophysiological processes causing general decline in the overall health of the afflicted population. The cumulative line of evidence suggests an important role of oxidative stress in the development and progression of the aging process and metabolic abnormalities, exacerbating adipocyte dysfunction, cardiovascular diseases, and associated complications at the same time. In recent years, robust have established the implication of Na/K-ATPase signaling in causing oxidative stress and alterations in cellular mechanisms, in addition to its distinct pumping function. Understanding the underlying molecular mechanisms and exploring the possible sources of pro-oxidants may allow for developing therapeutic targets in these processes and formulate novel intervention strategies for patients susceptible to aging and associated complications, such as obesity and cardiovascular disease. The attenuation of oxidative stress with targeted treatment options can improve patient outcomes and significantly reduce economic burden.
Collapse
|
40
|
Abstract
As aging involves oxidant injury, we examined the role of the recently described Na/K-ATPase oxidant amplification loop (NKAL). First, C57Bl6 old mice were given a western diet to stimulate oxidant injury or pNaKtide to antagonize the NKAL. The western diet accelerated functional and morphological evidence for aging whereas pNaKtide attenuated these changes. Next, human dermal fibroblasts (HDFs) were exposed to different types of oxidant stress in vitro each of which increased expression of senescence markers, cell-injury, and apoptosis as well as stimulated the NKAL. Further stimulation of the NKAL with ouabain augmented cellular senescence whereas treatment with pNaKtide attenuated it. Although N-Acetyl Cysteine and Vitamin E also ameliorated overall oxidant stress to a similar degree as pNaKtide, the pNaKtide produced protection against senescence that was substantially greater than that seen with either antioxidant. In particular, pNaKtide appeared to specifically ameliorate nuclear oxidant stress to a greater degree. These data demonstrate that the NKAL is intimately involved in the aging process and may serve as a target for anti-aging interventions.
Collapse
|
41
|
Drummond CA, Fan X, Haller ST, Kennedy DJ, Liu J, Tian J. Na/K-ATPase signaling mediates miR-29b-3p regulation and cardiac fibrosis formation in mice with chronic kidney disease. PLoS One 2018; 13:e0197688. [PMID: 29775473 PMCID: PMC5959191 DOI: 10.1371/journal.pone.0197688] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/07/2018] [Indexed: 01/19/2023] Open
Abstract
The Na/K-ATPase is an important membrane ion transporter and a signaling receptor that is essential for maintaining normal cell function. The current study examined the role of Na/K-ATPase signaling in regulating miR-29b-3p, an anti-fibrotic microRNA, in a mouse chronic kidney disease (CKD) model (5/6th partial nephrectomy or PNx). The results showed that CKD induced significant reduction of miR-29b-3p expression in the heart tissue by activation of Src and NFκB signaling in these animals. To demonstrate the role of Na/K-ATPase signaling, we also performed the PNx surgery on Na/K-ATPase α1 heterozygous (α1+/-) mice, which expresses ~40% less Na/K-ATPase α1 compared to their wild type littermates (WT) and exhibits deficiency in Na/K-ATPase signaling. We found that CKD did not significantly change the miR-29b-3p expression in heart tissue from the α1+/- animals. We also found that CKD failed to activate Src and NFκB signaling in these animals. Using isolated cardiac fibroblasts from α1+/- mice and their WT littermates, we showed that ouabain, a specific Na/K-ATPase ligand, induces decreased miR-29b-3p expression in fibroblasts isolated from WT mice, but had no effect in cells from α1+/- mice. Inhibition of NFκB by Bay11-7082 prevented ouabain-induced miR-29b-3p reduction in WT fibroblasts. To further confirm the in vivo effect of Na/K-ATPase signaling in regulation of miR-29b-3p and cardiac fibrosis in CKD animals, we used pNaKtide, a Src inhibiting peptide derived from the sequence of Na/K-ATPase, to block the activation of Na/K-ATPase signaling. The result showed that pNaKtide injection significantly increased miR-29b-3p expression and mitigated the CKD-induced cardiac fibrosis in these animals. These results clearly demonstrated that Na/K-ATPase signaling is an important mediator in CKD that regulates miR-29b-3p expression and cardiac fibrosis, which provides a novel target for regulation of miR-29b-3p in CKD. We also demonstrate that antagonizing Na/K-ATPase signaling by pNaKtide can reduce organ fibrosis through the stimulation of tissue miR-29b-3p expression.
Collapse
Affiliation(s)
| | - Xiaoming Fan
- Department of Medicine at the University of Toledo, Toledo, OH, United States of America
| | - Steven T. Haller
- Department of Medicine at the University of Toledo, Toledo, OH, United States of America
| | - David J. Kennedy
- Department of Medicine at the University of Toledo, Toledo, OH, United States of America
| | - Jiang Liu
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States of America
| | - Jiang Tian
- Department of Medicine at the University of Toledo, Toledo, OH, United States of America
- * E-mail:
| |
Collapse
|
42
|
Cheng X, Song Y, Wang Y. pNaKtide ameliorates renal interstitial fibrosis through inhibition of sodium-potassium adenosine triphosphatase-mediated signaling pathways in unilateral ureteral obstruction mice. Nephrol Dial Transplant 2018; 34:242-252. [DOI: 10.1093/ndt/gfy107] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/17/2018] [Indexed: 01/02/2023] Open
Affiliation(s)
- Xi Cheng
- Renal Division, Department of Medicine, Peking University First Hospital
- Institute of Nephrology, Peking University
- Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education
| | - Yi Song
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Yu Wang
- Renal Division, Department of Medicine, Peking University First Hospital
- Institute of Nephrology, Peking University
- Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education
| |
Collapse
|
43
|
Liu J, Lilly MN, Shapiro JI. Targeting Na/K-ATPase Signaling: A New Approach to Control Oxidative Stress. Curr Pharm Des 2018; 24:359-364. [PMID: 29318961 PMCID: PMC6052765 DOI: 10.2174/1381612824666180110101052] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 12/27/2017] [Accepted: 01/04/2017] [Indexed: 01/13/2023]
Abstract
Renal and cardiac function are greatly affected by chronic oxidative stress which can cause many pathophysiological states. The Na/K-ATPase is well-described as an ion pumping enzyme involved in maintaining cellular ion homeostasis; however, in the past two decades, extensive research has been done to understand the signaling function of the Na/K-ATPase and determine its role in physiological and pathophysiological states. Our lab has shown that the Na/K-ATPase signaling cascade can function as an amplifier of reactive oxygen species (ROS) which can be initiated by cardiotonic steroids or increases in ROS. Regulation of systemic oxidative stress by targeting Na/K-ATPase signaling mediated oxidant amplification improves 5/6th partial nephrectomy (PNx) mediated uremic cardiomyopathy, renal sodium handling, as well as ameliorates adipogenesis. This review will present this new concept of Na/K-ATPase signaling mediated oxidant amplification loop and its clinic implication.
Collapse
Affiliation(s)
- Jiang Liu
- Dept. of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Megan N. Lilly
- Dept. of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Joseph I. Shapiro
- Dept. of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| |
Collapse
|
44
|
Cui X, Xie Z. Protein Interaction and Na/K-ATPase-Mediated Signal Transduction. Molecules 2017; 22:molecules22060990. [PMID: 28613263 PMCID: PMC6152704 DOI: 10.3390/molecules22060990] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 02/05/2023] Open
Abstract
The Na/K-ATPase (NKA), or Na pump, is a member of the P-type ATPase superfamily. In addition to pumping ions across cell membrane, it is engaged in assembly of multiple protein complexes in the plasma membrane. This assembly allows NKA to perform many non-pumping functions including signal transduction that are important for animal physiology and disease progression. This article will focus on the role of protein interaction in NKA-mediated signal transduction, and its potential utility as target for developing new therapeutics.
Collapse
Affiliation(s)
- Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| |
Collapse
|
45
|
Zhang D, Zhang P, Yang P, He Y, Wang X, Yang Y, Zhu H, Xu N, Liang S. Downregulation of ATP1A1 promotes cancer development in renal cell carcinoma. Clin Proteomics 2017; 14:15. [PMID: 28484360 PMCID: PMC5418755 DOI: 10.1186/s12014-017-9150-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/26/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Aberrant expression of Na+/K+-ATPase α1 subunit (ATP1A1) is widely observed in multiple types of tumors, and its tissue-specific expression relates to cancer development. However, the functions and molecular mechanisms in renal cell carcinoma (RCC) are not fully understood. METHODS We investigated the ATP1A1 expression changes and possible roles in RCC through a quantitative proteomic approach and an integrative biochemical assessment. We detected ATP1A1 in RCC with LC-MS/MS, and further validated its expression with immunohistochemical analyses of 80 pairs of the RCC tumor and non-tumor tissues samples. The association of ATP1A1 expression with RCC pathology was statistically analyzed. Cell proliferation, migration and apoptosis were measured by CCK-8, boyden chamber assay and flow cytometry, respectively. The production of reactive oxygen species (ROS) was labeled with a single staining using a commercial kit, and was further detected with flow cytometry. RESULTS The ATP1A1 shows a significantly decreased expression in human RCC tissues than in the adjacent non-tumor tissues. The RCC patients with ATP1A1-positive expression exhibit longer overall survival time than the ATP1A1-negative patients. The exogenous overexpression of ATP1A1 inhibits RCC cell proliferation and cell migration by increasing the production of ROS. In addition, ATP1A1-mediated Raf/MEK/ERK signaling pathway is suppressed in RCC cells, indicating the possible occurrence of induced cell apoptosis. CONCLUSIONS Our in vitro and in vivo data of ATP1A1 inhibitory roles in RCC progression suggest that ATP1A1 is a potential novel suppressor protein for renal cancer.
Collapse
Affiliation(s)
- Dan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17 Section 3, People’s South Road, Chengdu, 610041 People’s Republic of China
| | - Peng Zhang
- Department of Urinary Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041 People’s Republic of China
| | - Pengbo Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17 Section 3, People’s South Road, Chengdu, 610041 People’s Republic of China
| | - Yu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17 Section 3, People’s South Road, Chengdu, 610041 People’s Republic of China
| | - Xixi Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17 Section 3, People’s South Road, Chengdu, 610041 People’s Republic of China
| | - Yanfang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17 Section 3, People’s South Road, Chengdu, 610041 People’s Republic of China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100021 People’s Republic of China
| | - Ningzhi Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17 Section 3, People’s South Road, Chengdu, 610041 People’s Republic of China
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100021 People’s Republic of China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17 Section 3, People’s South Road, Chengdu, 610041 People’s Republic of China
| |
Collapse
|
46
|
On the Many Actions of Ouabain: Pro-Cystogenic Effects in Autosomal Dominant Polycystic Kidney Disease. Molecules 2017; 22:molecules22050729. [PMID: 28467389 PMCID: PMC5688955 DOI: 10.3390/molecules22050729] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/19/2017] [Accepted: 04/30/2017] [Indexed: 02/07/2023] Open
Abstract
Ouabain and other cardenolides are steroidal compounds originally discovered in plants. Cardenolides were first used as poisons, but after finding their beneficial cardiotonic effects, they were rapidly included in the medical pharmacopeia. The use of cardenolides to treat congestive heart failure remained empirical for centuries and only relatively recently, their mechanisms of action became better understood. A breakthrough came with the discovery that ouabain and other cardenolides exist as endogenous compounds that circulate in the bloodstream of mammals. This elevated these compounds to the category of hormones and opened new lines of investigation directed to further study their biological role. Another important discovery was the finding that the effect of ouabain was mediated not only by inhibition of the activity of the Na,K-ATPase (NKA), but by the unexpected role of NKA as a receptor and a signal transducer, which activates a complex cascade of intracellular second messengers in the cell. This broadened the interest for ouabain and showed that it exerts actions that go beyond its cardiotonic effect. It is now clear that ouabain regulates multiple cell functions, including cell proliferation and hypertrophy, apoptosis, cell adhesion, cell migration, and cell metabolism in a cell and tissue type specific manner. This review article focuses on the cardenolide ouabain and discusses its various in vitro and in vivo effects, its role as an endogenous compound, its mechanisms of action, and its potential use as a therapeutic agent; placing especial emphasis on our findings of ouabain as a pro-cystogenic agent in autosomal dominant polycystic kidney disease (ADPKD).
Collapse
|
47
|
Wang X, Liu J, Drummond CA, Shapiro JI. Sodium potassium adenosine triphosphatase (Na/K-ATPase) as a therapeutic target for uremic cardiomyopathy. Expert Opin Ther Targets 2017; 21:531-541. [PMID: 28338377 PMCID: PMC5590225 DOI: 10.1080/14728222.2017.1311864] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/23/2017] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Clinically, patients with significant reductions in renal function present with cardiovascular dysfunction typically termed, uremic cardiomyopathy. It is a progressive series of cardiac pathophysiological changes, including left ventricular diastolic dysfunction and hypertrophy (LVH) which sometimes progress to left ventricular dilation (LVD) and systolic dysfunction in the setting of chronic kidney disease (CKD). Uremic cardiomyopathy is almost ubiquitous in patients afflicted with end stage renal disease (ESRD). Areas covered: This article reviews recent epidemiology, pathophysiology of uremic cardiomyopathy and provide a board overview of Na/K-ATPase research with detailed discussion on the mechanisms of Na/K-ATPase/Src/ROS amplification loop. We also present clinical and preclinical evidences as well as molecular mechanism of this amplification loop in the development of uremic cardiomyopathy. A potential therapeutic peptide that targets on this loop is discussed. Expert opinion: Current clinical treatment for uremic cardiomyopathy remains disappointing. Targeting the ROS amplification loop mediated by the Na/K-ATPase signaling function may provide a novel therapeutic target for uremic cardiomyopathy and related diseases. Additional studies of Na/K-ATPase and other strategies that regulate this loop will lead to new therapeutics.
Collapse
Affiliation(s)
- Xiaoliang Wang
- a Joan C Edwards School of Medicine at Marshall University , Huntington , WV , United States
- b University of Toledo College of Medicine and Life Sciences , Toledo , OH , United States
| | - Jiang Liu
- a Joan C Edwards School of Medicine at Marshall University , Huntington , WV , United States
| | - Christopher A Drummond
- b University of Toledo College of Medicine and Life Sciences , Toledo , OH , United States
| | - Joseph I Shapiro
- a Joan C Edwards School of Medicine at Marshall University , Huntington , WV , United States
| |
Collapse
|
48
|
Venugopal J, McDermott J, Sanchez G, Sharma M, Barbosa L, Reif GA, Wallace DP, Blanco G. Ouabain promotes partial epithelial to mesenchymal transition (EMT) changes in human autosomal dominant polycystic kidney disease (ADPKD) cells. Exp Cell Res 2017; 355:142-152. [PMID: 28385574 DOI: 10.1016/j.yexcr.2017.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/31/2017] [Accepted: 04/01/2017] [Indexed: 12/13/2022]
Abstract
The hormone ouabain has been shown to enhance the cystic phenotype of autosomal dominant polycystic kidney disease (ADPKD). Among other characteristics, the ADPKD phenotype includes cell de-differentiation and epithelial to mesenchymal transition (EMT). Here, we determined whether physiological concentrations of ouabain induces EMT in human renal epithelial cells from patients with ADPKD. We found that ADPKD cells respond to ouabain with a decrease in expression of the epithelial marker E-cadherin and increase in the expression of the mesenchymal markers N-cadherin, α smooth muscle actin (αSMA) and collagen-I; and the tight junction protein occludin and claudin-1. Other adhesion molecules, such as ZO-1, β-catenin and vinculin were not significantly modified by ouabain. At the cellular level, ouabain stimulated ADPKD cell migration, reduced cell-cell interaction, and the ability of ADPKD cells to form aggregates. Moreover, ouabain increased the transepithelial electrical resistance of ADPKD cell monolayers, suggesting that the paracellular transport pathway was preserved in the cells. These effects of ouabain were not observed in normal human kidney (NHK) cells. Altogether these results show a novel role for ouabain in ADPKD, inducing changes that lead to a partial EMT phenotype in the cells. These effects further support the key role that ouabain has as a factor that promotes the cystic characteristics of ADPKD cells.
Collapse
Affiliation(s)
- Jessica Venugopal
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Jeffrey McDermott
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Gladis Sanchez
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Madhulika Sharma
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Leandro Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Divinopolis, Brazil
| | - Gail A Reif
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Darren P Wallace
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States; Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Gustavo Blanco
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States.
| |
Collapse
|
49
|
Hangaard L, Bouzinova EV, Staehr C, Dam VS, Kim S, Xie Z, Aalkjaer C, Matchkov VV. Na-K-ATPase regulates intercellular communication in the vascular wall via cSrc kinase-dependent connexin43 phosphorylation. Am J Physiol Cell Physiol 2017; 312:C385-C397. [DOI: 10.1152/ajpcell.00347.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/10/2017] [Accepted: 01/14/2017] [Indexed: 12/23/2022]
Abstract
Communication between vascular smooth muscle cells (VSMCs) is dependent on gap junctions and is regulated by the Na-K-ATPase. The Na-K-ATPase is therefore important for synchronized VSMC oscillatory activity, i.e., vasomotion. The signaling between the Na-K-ATPase and gap junctions is unknown. We tested here the hypothesis that this signaling involves cSrc kinase. Intercellular communication was assessed by membrane capacitance measurements of electrically coupled VSMCs. Vasomotion in isometric myograph, input resistance, and synchronized [Ca2+]i transients were used as readout for intercellular coupling in rat mesenteric small arteries in vitro. Phosphorylation of cSrc kinase and connexin43 (Cx43) were semiquantified by Western blotting. Micromole concentration of ouabain reduced the amplitude of norepinephrine-induced vasomotion and desynchronized Ca2+ transients in VSMC in the arterial wall. Ouabain also increased input resistance in the arterial wall. These effects of ouabain were antagonized by inhibition of tyrosine phosphorylation with genistein, PP2, and by an inhibitor of the Na-K-ATPase-dependent cSrc activation, pNaKtide. Moreover, inhibition of cSrc phosphorylation increased vasomotion amplitude and decreased the resistance between cells in the vascular wall. Ouabain inhibited the electrical coupling between A7r5 cells, but pNaKtide restored the electrical coupling. Ouabain increased cSrc autophosphorylation of tyrosine 418 (Y418) required for full catalytic activity whereas pNaKtide antagonized it. This cSrc activation was associated with Cx43 phosphorylation of tyrosine 265 (Y265). Our findings demonstrate that Na-K-ATPase regulates intercellular communication in the vascular wall via cSrc-dependent Cx43 tyrosine phosphorylation.
Collapse
Affiliation(s)
- Lise Hangaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Vibeke S. Dam
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Sukhan Kim
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Christian Aalkjaer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, University of Copenhagen, Copenhagen, Denmark; and
| | | |
Collapse
|
50
|
Sodhi K, Srikanthan K, Goguet-Rubio P, Nichols A, Mallick A, Nawab A, Martin R, Shah PT, Chaudhry M, Sigdel S, El-Hamdani M, Liu J, Xie Z, Abraham NG, Shapiro JI. pNaKtide Attenuates Steatohepatitis and Atherosclerosis by Blocking Na/K-ATPase/ROS Amplification in C57Bl6 and ApoE Knockout Mice Fed a Western Diet. Sci Rep 2017; 7:193. [PMID: 28298638 PMCID: PMC5428305 DOI: 10.1038/s41598-017-00306-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 02/20/2017] [Indexed: 02/07/2023] Open
Abstract
We have previously reported that the α1 subunit of sodium potassium adenosine triphosphatase (Na/K-ATPase), acts as a receptor and an amplifier for reactive oxygen species, in addition to its distinct pumping function. On this background, we speculated that blockade of Na/K-ATPase-induced ROS amplification with a specific peptide, pNaKtide, might attenuate the development of steatohepatitis. To test this hypothesis, pNaKtide was administered to a murine model of NASH: the C57Bl6 mouse fed a “western” diet containing high amounts of fat and fructose. The administration of pNaKtide reduced obesity as well as hepatic steatosis, inflammation and fibrosis. Of interest, we also noted marked improvement in mitochondrial fatty acid oxidation, insulin sensitivity, dyslipidemia and aortic streaking in this mouse model. To further elucidate the effects of pNaKtide on atherosclerosis, similar studies were performed in ApoE knockout mice also exposed to the western diet. In these mice, pNaKtide not only improved steatohepatitis, dyslipidemia, and insulin sensitivity, but also ameliorated significant aortic atherosclerosis. Collectively, this study demonstrates that the Na/K-ATPase/ROS amplification loop contributes significantly to the development and progression of steatohepatitis and atherosclerosis. And furthermore, this study presents a potential treatment, the pNaKtide, for the metabolic syndrome phenotype.
Collapse
Affiliation(s)
- Komal Sodhi
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Krithika Srikanthan
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Perrine Goguet-Rubio
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Alexandra Nichols
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Amrita Mallick
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Athar Nawab
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Rebecca Martin
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Preeya T Shah
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Muhammad Chaudhry
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Saroj Sigdel
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Mehiar El-Hamdani
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Jiang Liu
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Zijian Xie
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA
| | - Nader G Abraham
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA.,Department of Medicine, New York Medical College, Valhalla, NY, 10595, USA
| | - Joseph I Shapiro
- Departments of Medicine, Surgery, Pathology, and Cardiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, USA.
| |
Collapse
|