1
|
Gao Y, Luo Y, Ji G, Wu T. Functional and pathological roles of adenylyl cyclases in various diseases. Int J Biol Macromol 2024; 281:136198. [PMID: 39366614 DOI: 10.1016/j.ijbiomac.2024.136198] [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: 07/15/2024] [Revised: 09/29/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
Adenylyl cyclases (ADCYs) produce the second messengers cAMP, which is crucial for a number of cellular activities. There are ten isoforms in the mammalian ADCY family including nine transmembrane adenylyl cyclases (tmAC) and one soluble adenylyl cyclase (sAC/ADCY10). There have been numerous studies demonstrating the importance of ADCYs in the development of a wide range of diseases, including cardiovascular disease, neurological disease, liver disease, and tumors. The classification, structure and regulation of ADCYs are discussed in this overview, which is followed by an analysis of how ADCYs are involved in various disorders and how they are used as a therapeutic tool. Our objective is to get a more thorough understanding of ADCYs to aid future study and provide novel ideas for the treatment of particular diseases.
Collapse
Affiliation(s)
- Ying Gao
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yanqun Luo
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Alzahrani NA, Bahaidrah KA, Mansouri RA, Aldhahri RS, Abd El-Aziz GS, Alghamdi BS. Possible Prophylactic Effects of Sulforaphane on LPS-Induced Recognition Memory Impairment Mediated by Regulating Oxidative Stress and Neuroinflammatory Proteins in the Prefrontal Cortex Region of the Brain. Biomedicines 2024; 12:1107. [PMID: 38791068 PMCID: PMC11118062 DOI: 10.3390/biomedicines12051107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/31/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) presents a significant global health concern, characterized by neurodegeneration and cognitive decline. Neuroinflammation is a crucial factor in AD development and progression, yet effective pharmacotherapy remains elusive. Sulforaphane (SFN), derived from cruciferous vegetables and mainly from broccoli, has shown a promising effect via in vitro and in vivo studies as a potential treatment for AD. This study aims to investigate the possible prophylactic mechanisms of SFN against prefrontal cortex (PFC)-related recognition memory impairment induced by lipopolysaccharide (LPS) administration. METHODOLOGY Thirty-six Swiss (SWR/J) mice weighing 18-25 g were divided into three groups (n = 12 per group): a control group (vehicle), an LPS group (0.75 mg/kg of LPS), and an LPS + SFN group (25 mg/kg of SFN). The total duration of the study was 3 weeks, during which mice underwent treatments for the initial 2 weeks, with daily monitoring of body weight and temperature. Behavioral assessments via novel object recognition (NOR) and temporal order recognition (TOR) tasks were conducted in the final week of the study. Inflammatory markers (IL-6 and TNF), antioxidant enzymes (SOD, GSH, and CAT), and pro-oxidant (MDA) level, in addition to acetylcholine esterase (AChE) activity and active (caspase-3) and phosphorylated (AMPK) levels, were evaluated. Further, PFC neuronal degeneration, Aβ content, and microglial activation were also examined using H&E, Congo red staining, and Iba1 immunohistochemistry, respectively. RESULTS SFN pretreatment significantly improved recognition memory performance during the NOR and TOR tests. Moreover, SFN was protected from neuroinflammation and oxidative stress as well as neurodegeneration, Aβ accumulation, and microglial hyperactivity. CONCLUSION The obtained results suggested that SFN has a potential protective property to mitigate the behavioral and biochemical impairments induced by chronic LPS administration and suggested to be via an AMPK/caspase-3-dependent manner.
Collapse
Affiliation(s)
- Noor Ahmed Alzahrani
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 23218, Saudi Arabia; (K.A.B.); (R.A.M.); (R.S.A.)
| | - Khulud Abdullah Bahaidrah
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 23218, Saudi Arabia; (K.A.B.); (R.A.M.); (R.S.A.)
| | - Rasha A. Mansouri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 23218, Saudi Arabia; (K.A.B.); (R.A.M.); (R.S.A.)
| | - Rahaf Saeed Aldhahri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 23218, Saudi Arabia; (K.A.B.); (R.A.M.); (R.S.A.)
- Department of Biochemistry, Faculty of Sciences, University of Jeddah, Jeddah 23218, Saudi Arabia
| | - Gamal S. Abd El-Aziz
- Department of Clinical Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia;
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
3
|
Patil NY, Rus I, Joshi AD. Role of ERK1/2 Signaling in Cinnabarinic Acid-Driven Stanniocalcin 2-Mediated Protection against Alcohol-Induced Apoptosis. J Pharmacol Exp Ther 2023; 387:111-120. [PMID: 37562971 PMCID: PMC10519581 DOI: 10.1124/jpet.123.001670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/12/2023] Open
Abstract
We have previously shown that a bona fide aryl hydrocarbon receptor (AhR) agonist, cinnabarinic acid (CA), protects against alcohol-induced hepatocyte apoptosis via activation of a novel AhR target gene, stanniocalcin 2 (Stc2). Stc2 translates to a secreted disulfide-linked hormone, STC2, known to function in cell development, calcium and phosphate regulation, angiogenesis, and antiapoptosis-albeit the comprehensive mechanism by which the CA-AhR-STC2 axis confers antiapoptosis is yet to be characterized. In this study, using RNA interference library screening, downstream antiapoptotic molecular signaling components involved in CA-induced STC2-mediated protection against ethanol-induced apoptosis were investigated. RNA interference library screening of kinases and phosphatases in Hepa1 cells and subsequent pathway analysis identified mitogen-activated protein kinase (MAPK) signaling as a critical molecular pathway involved in CA-mediated protection. Specifically, phosphorylation of ERK1/2 was induced in response to CA treatment without alterations in p38 and JNK signaling pathways. Silencing Stc2 in Hepa1 cells and in vivo experiments performed in Stc2-/- (Stc2 knockout) mice, which failed to confer CA-mediated protection against ethanol-induced apoptosis, showed abrogation of ERK1/2 activation, underlining the significance of ERK1/2 signaling in CA-STC2-mediated protection. In conclusion, activation of ERK1/2 signaling in CA-driven AhR-dependent Stc2-mediated protection represents a novel mechanism of protection against acute alcohol-induced apoptosis. SIGNIFICANCE STATEMENT: Previous studies have shown the role of stanniocalcin 2 (Stc2) in cinnabarinic acid (CA)-mediated protection against alcohol-induced apoptosis. Here, using RNA interference library screening and subsequent in vivo studies, the functional significance of ERK1/2 activation in CA-induced Stc2-mediated protection against acute ethanol-induced apoptosis was identified. This study is thus significant as it illustrates a comprehensive downstream mechanism by which CA-induced Stc2 protects against alcoholic liver disease.
Collapse
Affiliation(s)
- Nikhil Y Patil
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Iulia Rus
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Aditya D Joshi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
4
|
Salete-Granado D, Carbonell C, Puertas-Miranda D, Vega-Rodríguez VJ, García-Macia M, Herrero AB, Marcos M. Autophagy, Oxidative Stress, and Alcoholic Liver Disease: A Systematic Review and Potential Clinical Applications. Antioxidants (Basel) 2023; 12:1425. [PMID: 37507963 PMCID: PMC10376811 DOI: 10.3390/antiox12071425] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Ethanol consumption triggers oxidative stress by generating reactive oxygen species (ROS) through its metabolites. This process leads to steatosis and liver inflammation, which are critical for the development of alcoholic liver disease (ALD). Autophagy is a regulated dynamic process that sequesters damaged and excess cytoplasmic organelles for lysosomal degradation and may counteract the harmful effects of ROS-induced oxidative stress. These effects include hepatotoxicity, mitochondrial damage, steatosis, endoplasmic reticulum stress, inflammation, and iron overload. In liver diseases, particularly ALD, macroautophagy has been implicated as a protective mechanism in hepatocytes, although it does not appear to play the same role in stellate cells. Beyond the liver, autophagy may also mitigate the harmful effects of alcohol on other organs, thereby providing an additional layer of protection against ALD. This protective potential is further supported by studies showing that drugs that interact with autophagy, such as rapamycin, can prevent ALD development in animal models. This systematic review presents a comprehensive analysis of the literature, focusing on the role of autophagy in oxidative stress regulation, its involvement in organ-organ crosstalk relevant to ALD, and the potential of autophagy-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Daniel Salete-Granado
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
| | - Cristina Carbonell
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - David Puertas-Miranda
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Víctor-José Vega-Rodríguez
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Marina García-Macia
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Instituto de Biología Funcional y Genómica (IBFG), Universidad de Salamanca, 37007 Salamanca, Spain
| | - Ana Belén Herrero
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Miguel Marcos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
5
|
Li C, Hao J, Qiu H, Xin H. CaMKK2 alleviates myocardial ischemia/reperfusion injury by inhibiting oxidative stress and inflammation via the action on the AMPK-AKT-GSK-3β/Nrf2 signaling cascade. Inflamm Res 2023:10.1007/s00011-023-01756-6. [PMID: 37338678 DOI: 10.1007/s00011-023-01756-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/21/2023] [Accepted: 06/02/2023] [Indexed: 06/21/2023] Open
Abstract
OBJECTIVE Calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) can regulate numerous biological processes and is implicated in diverse pathological processes. Yet its role in myocardial ischemia/reperfusion (MI/R) injury remains unknown. This project explored the possible functions and mechanisms of CaMKK2 in MI/R injury. METHODS A rat model of MI/R in vivo was established using the left anterior descending coronary artery ligation method. Rat cardiomyocytes were exposed to hypoxia/reoxygenation (H/R) in vitro to establish a cell model. Overexpression of CaMKK2 was achieved by infecting recombinant adeno-associated virus or adenovirus expressing CaMKK2. Real-time quantitative PCR, immunoblotting, TTC staining, TUNEL assay, ELISA, oxidative stress detection assays, flow cytometry, and CCK-8 assay were carried out. RESULTS A decline in CaMKK2 levels was induced by MI/R in vivo or H/R in vitro. Up-modulation of CaMKK2 in rats ameliorated the cardiac injury evoked by MI/R injury accompanied by suppression of cardiac apoptosis, oxidative stress, and proinflammatory response. Rat cardiomyocytes with CaMKK2 overexpression were also protected from H/R damage by inhibiting apoptosis, oxidative stress, and proinflammatory response. CaMKK2 overexpression led to increased phosphorylation of AMPK, AKT, and GSK-3β, and enhanced activation of Nrf2 under MI/R or H/R conditions. Inhibition of AMPK abolished CaMKK2-mediated Nrf2 activation and relevant cardioprotective effect. Restraint of Nrf2 also diminished CaMKK2-mediated relevant cardioprotective effect. CONCLUSIONS Up-regulation of CaMKK2 provides a therapeutic benefit in the rat model of MI/R injury by boosting the Nrf2 pathway through regulation of AMPK/AKT/GSK-3β, which suggests CaMKK2 as a new molecular target for the treatment of MI/R injury.
Collapse
Affiliation(s)
- Chengliang Li
- Department of General Practice, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Jiajia Hao
- Department of General Practice, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Huichang Qiu
- Department of General Practice, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Hong Xin
- Healthcare Simulation Center, Department of Research, Education and Information, Guangzhou First People's Hospital, No.1 Panfu Road, Guangzhou, 510180, China.
| |
Collapse
|
6
|
Yan C, Hu W, Tu J, Li J, Liang Q, Han S. Pathogenic mechanisms and regulatory factors involved in alcoholic liver disease. J Transl Med 2023; 21:300. [PMID: 37143126 PMCID: PMC10158301 DOI: 10.1186/s12967-023-04166-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023] Open
Abstract
Alcoholism is a widespread and damaging behaviour of people throughout the world. Long-term alcohol consumption has resulted in alcoholic liver disease (ALD) being the leading cause of chronic liver disease. Many metabolic enzymes, including alcohol dehydrogenases such as ADH, CYP2E1, and CATacetaldehyde dehydrogenases ALDHsand nonoxidative metabolizing enzymes such as SULT, UGT, and FAEES, are involved in the metabolism of ethanol, the main component in alcoholic beverages. Ethanol consumption changes the functional or expression profiles of various regulatory factors, such as kinases, transcription factors, and microRNAs. Therefore, the underlying mechanisms of ALD are complex, involving inflammation, mitochondrial damage, endoplasmic reticulum stress, nitrification, and oxidative stress. Moreover, recent evidence has demonstrated that the gut-liver axis plays a critical role in ALD pathogenesis. For example, ethanol damages the intestinal barrier, resulting in the release of endotoxins and alterations in intestinal flora content and bile acid metabolism. However, ALD therapies show low effectiveness. Therefore, this review summarizes ethanol metabolism pathways and highly influential pathogenic mechanisms and regulatory factors involved in ALD pathology with the aim of new therapeutic insights.
Collapse
Affiliation(s)
- Chuyun Yan
- Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Wanting Hu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Key Lab of Microanalytical Methods & Instrumentation, Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
| | - Jinqi Tu
- The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College of Wuhu, Wannan Medical College, Wuhu, 241000, Anhui, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Key Lab of Microanalytical Methods & Instrumentation, Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
| | - Shuxin Han
- Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830046, China.
| |
Collapse
|
7
|
Hermes TDA, Mâncio RD, Mizobutti DS, Macedo AB, Kido LA, Cagnon Quitete VHA, Minatel E. Cilostazol attenuates oxidative stress and apoptosis in the quadriceps muscle of the dystrophic mouse experimental model. Int J Exp Pathol 2023; 104:13-22. [PMID: 36565167 PMCID: PMC9845609 DOI: 10.1111/iep.12461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/04/2022] [Accepted: 10/18/2022] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most severe and frequent form of muscular dystrophy. The mdx mouse is one of the most widely used experimental models to understand aspects of the biology of dystrophic skeletal muscles and the mechanisms of DMD. Oxidative stress and apoptosis are present in early stages of the disease in mdx mice. The high production of reactive oxygen species (ROS) causes activation of apoptotic death regulatory proteins due to DNA damage and breakdown of nuclear and mitochondrial membranes. The quadriceps (QUA) muscle of the mdx mouse is a good tool to study oxidative events. Previous studies have demonstrated that cilostazol exerts an anti-oxidant effect by decreasing the production of reactive oxygen species (ROS). The present study aimed to evaluate the ability of cilostazol to modulate oxidative stress and apoptosis in the QUA muscle of mdx mice. Fourteen-day-old mdx mice received cilostazol or saline for 14 days. C57BL/10 mice were used as a control. In the QUA muscle of mdx mice, cilostazol treatment decreased ROS production (-74%), the number of lipofuscin granules (-47%), lipid peroxidation (-11%), and the number of apoptotic cells (-66%). Thus cilostazol showed anti-oxidant and anti-apoptotic action in the QUA muscle of mdx mice.
Collapse
Affiliation(s)
- Túlio de Almeida Hermes
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
- Departament of Anatomy, Institute of Biomedical SciencesFederal University of Alfenas (UNIFAL‐MG)AlfenasBrazil
| | - Rafael Dias Mâncio
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
| | - Daniela Sayuri Mizobutti
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
| | - Aline Barbosa Macedo
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
| | - Larissa Akemi Kido
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
| | | | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
| |
Collapse
|
8
|
Rivas MV, Musikant D, Díaz Peña R, Álvarez D, Pelazzo L, Rossi E, Martínez KD, Errea MI, Pérez OE, Varela O, Kolender AA. Carbohydrate-Derived Polytriazole Nanoparticles Enhance the Anti-Inflammatory Activity of Cilostazol. ACS OMEGA 2022; 7:44631-44642. [PMID: 36530317 PMCID: PMC9753171 DOI: 10.1021/acsomega.2c02969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/10/2022] [Indexed: 06/17/2023]
Abstract
Poly(amide-triazole) and poly(ester-triazole) synthesized from d-galactose as a renewable resource were applied for the synthesis of nanoparticles (NPs) by the emulsification/solvent evaporation method. The NPs were characterized as stable, spherical particles, and none of their components, including the stabilizer poly(vinyl alcohol), were cytotoxic for normal rat kidney cells. These NPs proved to be useful for the efficient encapsulation of cilostazol (CLZ), an antiplatelet and vasodilator drug currently used for the treatment of intermittent claudication, which is associated with undesired side-effects. In this context, the nanoencapsulation of CLZ was expected to improve its therapeutic administration. The carbohydrate-derived polymeric NPs were designed taking into account that the triazole rings of the polymer backbone could have attractive interactions with the tetrazole ring of CLZ. The activity of the nanoencapsulated CLZ was measured using a matrix metalloproteinase model in a lipopolysaccharide-induced inflammation system. Interestingly, the encapsulated drug exhibited enhanced anti-inflammatory activity in comparison with the free drug. The results are very promising since the stable, noncytotoxic NP systems efficiently reduced the inflammation response at low CLZ doses. In summary, the NPs were obtained through an innovative methodology that combines a carbohydrate-derived synthetic polymer, designed to interact with the drug, ease of preparation, adequate biological performance, and environmentally friendly production.
Collapse
Affiliation(s)
- M. Verónica Rivas
- Universidad
de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
- Consejo
Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Centro de Investigación en Hidratos de Carbono
(CIHIDECAR), Ciudad Universitaria,
Pabellón 2, C1428EHABuenos Aires, Argentina
| | - Daniel Musikant
- Universidad
de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
- Consejo
Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Instituto de Química Biológica de la
Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
| | - Rocío Díaz Peña
- Universidad
de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
- Consejo
Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Instituto de Química Biológica de la
Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
| | - Daniela Álvarez
- Universidad
de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
- Consejo
Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Instituto de Química Biológica de la
Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
| | - Luciana Pelazzo
- Universidad
de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
- Consejo
Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Centro de Investigación en Hidratos de Carbono
(CIHIDECAR), Ciudad Universitaria,
Pabellón 2, C1428EHABuenos Aires, Argentina
| | - Ezequiel Rossi
- Instituto
Tecnológico de Buenos Aires (ITBA), Lavardén 315, C1437FBGBuenos Aires, Argentina
| | - Karina D. Martínez
- Facultad
de Arquitectura Diseño y Urbanismo, Universidad de Buenos Aires (UBA), Ciudad Universitaria, Pabellón 3, C1428EHABuenos Aires, Argentina
- Consejo Nacional
de Investigaciones Científicas y Técnicas (CONICET)-UBA, Instituto de Tecnología en Polímeros
y Nanotecnología (ITPN), Ciudad Universitaria, Pabellón 3, C1428EHABuenos Aires, Argentina
| | - María I. Errea
- Instituto
Tecnológico de Buenos Aires (ITBA), Lavardén 315, C1437FBGBuenos Aires, Argentina
| | - Oscar E. Pérez
- Universidad
de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
- Consejo
Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Instituto de Química Biológica de la
Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
| | - Oscar Varela
- Universidad
de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
- Consejo
Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Centro de Investigación en Hidratos de Carbono
(CIHIDECAR), Ciudad Universitaria,
Pabellón 2, C1428EHABuenos Aires, Argentina
| | - Adriana A. Kolender
- Universidad
de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria, Pabellón
2, C1428EHABuenos
Aires, Argentina
- Consejo
Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Centro de Investigación en Hidratos de Carbono
(CIHIDECAR), Ciudad Universitaria,
Pabellón 2, C1428EHABuenos Aires, Argentina
| |
Collapse
|
9
|
Kim YS, Ko B, Kim DJ, Tak J, Han CY, Cho JY, Kim W, Kim SG. Induction of the hepatic aryl hydrocarbon receptor by alcohol dysregulates autophagy and phospholipid metabolism via PPP2R2D. Nat Commun 2022; 13:6080. [PMID: 36241614 PMCID: PMC9568535 DOI: 10.1038/s41467-022-33749-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
Disturbed lipid metabolism precedes alcoholic liver injury. Whether and how AhR alters degradation of lipids, particularly phospho-/sphingo-lipids during alcohol exposure, was not explored. Here, we show that alcohol consumption in mice results in induction and activation of aryl hydrocarbon receptor (AhR) in the liver, and changes the hepatic phospho-/sphingo-lipids content. The levels of kynurenine, an endogenous AhR ligand, are elevated with increased hepatic tryptophan metabolic enzymes in alcohol-fed mice. Either alcohol or kynurenine treatment promotes AhR activation with autophagy dysregulation via AMPK. Protein Phosphatase 2 Regulatory Subunit-Bdelta (Ppp2r2d) is identified as a transcriptional target of AhR. Consequently, PPP2R2D-dependent AMPKα dephosphorylation causes autophagy inhibition and mitochondrial dysfunction. Hepatocyte-specific AhR ablation attenuates steatosis, which is associated with recovery of phospho-/sphingo-lipids content. Changes of AhR targets are corroborated using patient specimens. Overall, AhR induction by alcohol inhibits autophagy in hepatocytes through AMPKα, which is mediated by Ppp2r2d gene transactivation, revealing an AhR-dependent metabolism of phospho-/sphingo-lipids.
Collapse
Affiliation(s)
- Yun Seok Kim
- grid.31501.360000 0004 0470 5905Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, 03080 Korea ,grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea ,grid.31501.360000 0004 0470 5905College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Bongsub Ko
- grid.31501.360000 0004 0470 5905Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, 03080 Korea
| | - Da Jung Kim
- grid.31501.360000 0004 0470 5905Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, 03080 Korea ,grid.412484.f0000 0001 0302 820XMetabolomics Core Facility, Department of Transdisciplinary Research and Collaboration, Biomedical Research Institute, Seoul National University Hospital, Seoul, 03082 Korea
| | - Jihoon Tak
- grid.31501.360000 0004 0470 5905College of Pharmacy, Seoul National University, Seoul, Republic of Korea ,grid.255168.d0000 0001 0671 5021College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Kyeonggi-do 10326 Republic of Korea
| | - Chang Yeob Han
- grid.31501.360000 0004 0470 5905College of Pharmacy, Seoul National University, Seoul, Republic of Korea ,grid.411545.00000 0004 0470 4320School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, 54896 Korea
| | - Joo-Youn Cho
- grid.31501.360000 0004 0470 5905Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, 03080 Korea ,grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Won Kim
- grid.31501.360000 0004 0470 5905Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | - Sang Geon Kim
- grid.255168.d0000 0001 0671 5021College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Kyeonggi-do 10326 Republic of Korea
| |
Collapse
|
10
|
Wang Z, Liu H, Li L, Li Y, Yan H, Yuan Y. Modulation of Disordered Bile Acid Homeostasis and Hepatic Tight Junctions Using Salidroside against Hepatocyte Apoptosis in Furan-Induced Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10031-10043. [PMID: 35939816 DOI: 10.1021/acs.jafc.2c04654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Furan, a processing-induced food contaminant, has attracted great attention due to its hepatotoxicity. To further investigate the underlying mechanism of salidroside (SAL) alleviating furan-induced liver damage, we divided Balb/c mice into the control group, the furan (8 mg/kg/day) group, and three groups of three different doses of SAL (10/20/40 mg/kg/day) in the current research. The shifted serum profile was observed through untargeted metabonomics, to which the bile acid metabolism was related, and the alleviating effect of SAL against furan-induced apoptosis was caused by the metabolism. Target bile acid quantification for the liver and serum showed that SAL positively regulated the homeostasis of bile acids disturbed by furan. Meanwhile, SAL significantly upregulated the synthesis genes of bile acids (Cyp7a1, Cyp7b1, Cyp8b1, and Cyp27a1) and the uptake transport genes (Ntcp and Oatps) and downregulated the efflux transport genes (Bsep, Ost-α, Ost-β, Mrp2, and Mrp4). Transmission electron microscopy of the bile canaliculi and tight junctions and the levels of tight junction marker proteins (ZO-1, occludin, and claudin-1) confirmed that the disruption of the hepatic tight junction was inhibited by SAL. The connection between the apoptosis- and tight junction-related proteins was observed through the construction of a protein-protein interaction network. SAL suppressed the furan-induced hepatocyte apoptosis evidenced by the detection of TUNEL and Bax, Bcl-2, and caspase-3 levels. Taken together, SAL alleviated furan-induced hepatocyte apoptosis via altering the disordered homeostasis of bile acids and hepatic tight junctions.
Collapse
Affiliation(s)
- Ziyue Wang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Hui Liu
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Lu Li
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Yucai Li
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Haiyang Yan
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Yuan Yuan
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| |
Collapse
|
11
|
Naderbar L, Pazhang Y, Rezaie J. Inhibiting AKT signaling pathway with cilostazol and meloxicam synergism for suppressing K562 cells in vitro. J Biochem Mol Toxicol 2022; 36:e23185. [PMID: 35920412 DOI: 10.1002/jbt.23185] [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: 09/17/2021] [Revised: 05/30/2022] [Accepted: 07/25/2022] [Indexed: 11/11/2022]
Abstract
Despite advances in cancer treatment, chronic myeloid leukemia (CML) is still one of the leading causes of death in the world. Due to the role of inflammation in cancer promotion and progression, thus use of anti-inflammatory agents may suppress cancer cell growth. In this study, we used two anti-inflammatory drugs, cilostazol and meloxicam, for the treatment of CML. Cell viability was measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and the synergism occurrence was calculated by compusyn software. Annexin V/PI test and Hoechst staining were used to determine the apoptosis rate. To determine the pathway of apoptosis induction, the expression of BCL2 Associated X (Bax) and B-cell lymphoma-2 (Bcl-2) apoptotic genes and caspases activity were evaluated. The cell cycle was analyzed by propidium iodide (PI) staining and flow cytometry. Western blot analysis and immunofluorescence were performed to estimate alterations in Ak strain transforming-1 (AKT-1), phosphprylated AKT-1 (p-AKT-1), adenosine mono-phosphate-kinase (AMPK), and phosphorylated AMPK (p-AMPK) proteins and BCR/ABL and c-Myc distribution, respectively. Results showed that cilostazol, meloxicam, and their combination drug reduced cell viability (p < 0.05). Compared with control, expression of Bax and Bcl-2 decreased in treated cells, respectively (p < 0.05). The caspase-9 activity increased in treated cells compared to control cells (p < 0.001). The applied drugs decreased the protein level of p-AKT-1 while increasing the p-AMPK protein level (p < 0.05). BCR/ABL and c-Myc Protein distribution significantly decreased in treated cells. In conclusion, the combination drug had more cytotoxic effects than cilostazol and meloxicam alone and induced apoptosis by inhibiting AKT-1 activation and c-Myc reduction. Therefore using combination drugs effectively can treat cancers of CML origin.
Collapse
Affiliation(s)
- Laya Naderbar
- Biology department, Faculty of Sciences, Urmia University, Urmia, Iran
| | - Yaghub Pazhang
- Biology department, Faculty of Sciences, Urmia University, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
12
|
Xie F, Zhong Y, Wang D, So KF, Xiao J, Lv Y. Metformin protects against ethanol-induced liver triglyceride accumulation by the LKB1/AMPK/ACC pathway. Mol Biol Rep 2022; 49:7837-7848. [PMID: 35733070 DOI: 10.1007/s11033-022-07610-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: 02/14/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Hepatic lipid accumulation is one of the main pathological features of alcoholic liver disease (ALD). Metformin serves as an AMPK activator and has been shown to have lipids lowering effects in non-alcoholic fatty liver disease (NAFLD), but its role in ALD remains unclear. The purpose of this study was to explore the potential mechanism of metformin regulating lipid metabolism in ALD. METHODS AND RESULTS In vitro and in vivo ALD models were established using AML12 cells and C57BL/6 mice, respectively. To determine the effect of metformin on ALD in vitro, the concentration of cellular triglyceride was examined by Nile red staining and a biochemical kit. To further reveal the role of metformin on ALD in vivo, liver tissues were examined by HE and oil red O staining, and the levels of ALT and AST in serum were determined via an automatic biochemical analyzer. The expression of mRNA and proteins were measured using qRT-PCR and Western blot, respectively. The role of the LKB1/AMPK/ACC axis on metformin regulating ethanol-induced lipid accumulation was evaluated by siRNA and AAV-shRNA interference. The results showed metformin reduced the ethanol-induced lipid accumulation in AML12 cells through activating AMPK, inhibiting ACC, reducing SREBP1c, and increasing PPARα. In addition, compared with control mice, metformin treatment inhibited ethanol-induced liver triglyceride accumulation and the increase of ALT and AST in serum. Interference with LKB1 attenuated the effect of metformin on ethanol-induced lipid accumulation both in vitro and in vivo. CONCLUSION Metformin protects against lipid formation in ALD by activating the LKB1/AMPK/ACC axis.
Collapse
Affiliation(s)
- Fotian Xie
- Laboratory of Neuroendocrinology, Fujian Key Laboratory of Developmental and Neurobiology, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yuanming Zhong
- School of Physical Education and Sport Science, Fujian normal university, Fuzhou, China
| | - Dongmei Wang
- Laboratory of Neuroendocrinology, Fujian Key Laboratory of Developmental and Neurobiology, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Kwok Fai So
- Laboratory of Neuroendocrinology, Fujian Key Laboratory of Developmental and Neurobiology, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Jia Xiao
- Laboratory of Neuroendocrinology, Fujian Key Laboratory of Developmental and Neurobiology, College of Life Sciences, Fujian Normal University, Fuzhou, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yi Lv
- Laboratory of Neuroendocrinology, Fujian Key Laboratory of Developmental and Neurobiology, College of Life Sciences, Fujian Normal University, Fuzhou, China.
| |
Collapse
|
13
|
Lu J, Zhang Y, Wang YZ, Li YY, Wang R, Zhong YJ, Chen L, Song MW, Shi L, Li L, Li YW. Caffeic acid dimethyl ether alleviates alcohol-induced hepatic steatosis via microRNA-378b-mediated CaMKK2-AMPK pathway. Bioengineered 2022; 13:11122-11136. [PMID: 35481488 PMCID: PMC9208468 DOI: 10.1080/21655979.2022.2060586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Alcoholic liver disease (ALD), with its increasing morbidity and mortality, has seriously and extensively affected the health of people worldwide. Caffeic Acid Dimethyl Ether (CADE) significantly inhibits alcohol-induced hepatic steatosis in vivo through AMP-activated protein kinase (AMPK) pathway, but its in-depth mechanism remains unclear. This work aimed to clarify further mechanism of CADE in improving hepatic lipid accumulation in ALD through the microRNA-378b (miR-378b)-mediated Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2)-AMPK signaling pathway. Here, we reported that the hepatic or serum triglyceride (TG), total cholesterol (TC), alanine aminotransferase (ALT), and aspartate transaminase (AST) levels were sharply escalated by ethanol while prominently decreased by CADE. Ethanol sharply up-regulated miR-378b expression while CADE effectively prevented the elevation of miR-378b in vivo. And treatment of CADE surely increased mRNA and protein expression of CaMKK2 as a kinase of AMPK and reduced lipid accumulation in the livers of alcohol-fed C57BL/6 mice. MiR-378b escalation exacerbated hepatic steatosis and inhibited CaMKK2-AMPK signaling, while miR-378b deficiency alleviated lipid accumulation and activated the CaMKK2 cascade. Furthermore, CADE alleviated the lipid deposition and reversed the disorder of CaMKK2-AMPK signaling pathway induced by miR-378b over-expression. However, knockdown of miR-378b eliminated the beneficial effect of CADE on lipid metabolism. In brief, our results showed that CADE ultimately improved hepatic lipid deposition by regulating the CaMKK2-AMPK signaling pathway through miR-378b.
Collapse
Affiliation(s)
- Jun Lu
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Yan Zhang
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Ying-Zhao Wang
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Yuan-Yuan Li
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Rui Wang
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Yu-Juan Zhong
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Li Chen
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Meng-Wei Song
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Lin Shi
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Li Li
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Yong-Wen Li
- College of Pharmacy, Guilin Medical University, Guilin, China.,Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin, China
| |
Collapse
|
14
|
Sim KH, Shu MS, Kim S, Kim JY, Choi BH, Lee YJ. Cilostazol Induces Apoptosis and Inhibits Proliferation of Hepatocellular Carcinoma Cells by Activating AMPK. BIOTECHNOL BIOPROC E 2021. [DOI: 10.1007/s12257-021-0002-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
15
|
Lee H, Jiang X, Perwaiz I, Yu P, Wang J, Wang Y, Hüttemann M, Felder RA, Sibley DR, Polster BM, Rozyyev S, Armando I, Yang Z, Qu P, Jose PA. Dopamine D 5 receptor-mediated decreases in mitochondrial reactive oxygen species production are cAMP and autophagy dependent. Hypertens Res 2021; 44:628-641. [PMID: 33820956 PMCID: PMC8369611 DOI: 10.1038/s41440-021-00646-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 11/10/2019] [Accepted: 12/03/2019] [Indexed: 01/31/2023]
Abstract
Overproduction of reactive oxygen species (ROS) plays an important role in the pathogenesis of hypertension. The dopamine D5 receptor (D5R) is known to decrease ROS production, but the mechanism is not completely understood. In HEK293 cells overexpressing D5R, fenoldopam, an agonist of the two D1-like receptors, D1R and D5R, decreased the production of mitochondria-derived ROS (mito-ROS). The fenoldopam-mediated decrease in mito-ROS production was mimicked by Sp-cAMPS but blocked by Rp-cAMPS. In human renal proximal tubule cells with DRD1 gene silencing to eliminate the confounding effect of D1R, fenoldopam still decreased mito-ROS production. By contrast, Sch23390, a D1R and D5R antagonist, increased mito-ROS production in the absence of D1R, D5R is constitutively active. The fenoldopam-mediated inhibition of mito-ROS production may have been related to autophagy because fenoldopam increased the expression of the autophagy hallmark proteins, autophagy protein 5 (ATG5), and the microtubule-associated protein 1 light chain (LC)3-II. In the presence of chloroquine or spautin-1, inhibitors of autophagy, fenoldopam further increased ATG5 and LC3-II expression, indicating an important role of D5R in the positive regulation of autophagy. However, when autophagy was inhibited, fenoldopam was unable to inhibit ROS production. Indeed, the levels of these autophagy hallmark proteins were decreased in the kidney cortices of Drd5-/- mice. Moreover, ROS production was increased in mitochondria isolated from the kidney cortices of Drd5-/- mice, relative to Drd5+/+ littermates. In conclusion, D5R-mediated activation of autophagy plays a role in the D5R-mediated inhibition of mito-ROS production in the kidneys.
Collapse
Affiliation(s)
- Hewang Lee
- Department of Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA,Institute of Heart and Vessel Diseases, Affiliated Second Hospital, Dalian Medical University, Dalian, China,Division of Nephrology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA,Center for Molecular Physiology Research, Children’s Research Institute, Children’s National Medical Center, Washington, DC, USA,Department of Pediatrics, Georgetown University Medical Center, Washington, DC, USA,Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Xiaoliang Jiang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Imran Perwaiz
- Institute of Heart and Vessel Diseases, Affiliated Second Hospital, Dalian Medical University, Dalian, China
| | - Peiying Yu
- Division of Nephrology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA,Center for Molecular Physiology Research, Children’s Research Institute, Children’s National Medical Center, Washington, DC, USA,Department of Pediatrics, Georgetown University Medical Center, Washington, DC, USA
| | - Jin Wang
- Institute of Heart and Vessel Diseases, Affiliated Second Hospital, Dalian Medical University, Dalian, China
| | - Ying Wang
- Institute of Heart and Vessel Diseases, Affiliated Second Hospital, Dalian Medical University, Dalian, China
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics and Cardiovascular Research Institute, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Robin A. Felder
- Department of Pathology, University of Virginia Health Sciences Center, Charlottesville, VA, USA
| | - David R. Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Brian M. Polster
- Department of Anesthesiology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Selim Rozyyev
- Department of Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA
| | - Ines Armando
- Department of Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA,Division of Nephrology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA,Center for Molecular Physiology Research, Children’s Research Institute, Children’s National Medical Center, Washington, DC, USA,Department of Pediatrics, Georgetown University Medical Center, Washington, DC, USA
| | - Zhiwei Yang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Peng Qu
- Institute of Heart and Vessel Diseases, Affiliated Second Hospital, Dalian Medical University, Dalian, China
| | - Pedro A. Jose
- Department of Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA,Division of Nephrology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA,Center for Molecular Physiology Research, Children’s Research Institute, Children’s National Medical Center, Washington, DC, USA,Department of Pediatrics, Georgetown University Medical Center, Washington, DC, USA,Department of Pharmacology and Physiology, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA
| |
Collapse
|
16
|
Zhou Q, Wang L, Liu B, Xiao J, Cheng KW, Chen F, Wang M. Tricoumaroylspermidine from rose exhibits inhibitory activity against ethanol-induced apoptosis in HepG2 cells. Food Funct 2021; 12:5892-5902. [PMID: 34019608 DOI: 10.1039/d1fo00800e] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Hepatocyte apoptosis is involved in the pathogenesis of alcohol-associated liver disease (ALD) and anti-apoptotic agents/extracts are thereby of great importance in the prevention/treatment of ALD. In this study, the protective effects of 10 edible flowers against ethanol-induced cell death were investigated in HepG2 cells, with rose (Rosa rugosa) showing the strongest activity. Therefore, rose was chosen for further separation and purification of bioactive fractions. A special fraction, SLs, was found to significantly increase the viability of EtOH-treated cells and attenuated EtOH-induced apoptosis partially via the activation of the AMPK/SIRT1 signaling pathway. Chromatographic analysis identified a series of hydroxycinnamic acid amides, kaempferol glycosides, and quercetin glycosides in this fraction, while the following intracellular uptake and cytotoxicity studies revealed that N1,N5,N10-(E)-tri-p-coumaroylspermidine (a hydroxycinnamic acid amide) in this fraction exhibited remarkable hepatoprotective activity with similar effective dosage to sulforaphane. Hence, our results highlighted the anti-alcohol and hepatoprotective benefits of consuming rose.
Collapse
Affiliation(s)
- Qian Zhou
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China. and Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| | - Lanxiang Wang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China and Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bin Liu
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China. and Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| | - Jianbo Xiao
- Institute of Food Safety and Nutrition, Jiangsu University, Zhenjiang, China and Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo, Vigo, Spain
| | - Ka-Wing Cheng
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China. and Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| | - Feng Chen
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China. and Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| | - Mingfu Wang
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China. and Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China and School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
17
|
cAMP Signaling in Pathobiology of Alcohol Associated Liver Disease. Biomolecules 2020; 10:biom10101433. [PMID: 33050657 PMCID: PMC7600246 DOI: 10.3390/biom10101433] [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: 09/21/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023] Open
Abstract
The importance of cyclic adenosine monophosphate (cAMP) in cellular responses to extracellular signals is well established. Many years after discovery, our understanding of the intricacy of cAMP signaling has improved dramatically. Multiple layers of regulation exist to ensure the specificity of cellular cAMP signaling. Hence, disturbances in cAMP homeostasis could arise at multiple levels, from changes in G protein coupled receptors and production of cAMP to the rate of degradation by phosphodiesterases. cAMP signaling plays critical roles in metabolism, inflammation and development of fibrosis in several tissues. Alcohol-associated liver disease (ALD) is a multifactorial condition ranging from a simple steatosis to steatohepatitis and fibrosis and ultimately cirrhosis, which might lead to hepatocellular cancer. To date, there is no FDA-approved therapy for ALD. Hence, identifying the targets for the treatment of ALD is an important undertaking. Several human studies have reported the changes in cAMP homeostasis in relation to alcohol use disorders. cAMP signaling has also been extensively studied in in vitro and in vivo models of ALD. This review focuses on the role of cAMP in the pathobiology of ALD with emphasis on the therapeutic potential of targeting cAMP signaling for the treatment of various stages of ALD.
Collapse
|
18
|
Siregar AS, Nyiramana MM, Kim EJ, Shin EJ, Woo MS, Kim JM, Kim JH, Lee DK, Hahm JR, Kim HJ, Kim CW, Kim NG, Park SH, Choi YJ, Kang SS, Hong SG, Han J, Kang D. Dipeptide YA is Responsible for the Positive Effect of Oyster Hydrolysates on Alcohol Metabolism in Single Ethanol Binge Rodent Models. Mar Drugs 2020; 18:md18100512. [PMID: 33050644 PMCID: PMC7601867 DOI: 10.3390/md18100512] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
Accumulative alcohol hangovers cause liver damage through oxidative and inflammatory stress. Numerous antioxidant and anti-inflammatory reagents have been developed to reduce alcohol hangovers, but these reagents are still insignificant and have limitations in that they can cause liver toxicity. Oyster hydrolysate (OH), another reagent that has antioxidant and anti-inflammatory activity, is a product extracted through an enzymatic hydrolysis process from oysters (Crassostrea gigas), which can be easily eaten in meals. This study was aimed at determining the effects of OH on alcohol metabolism, using a single high dose of ethanol (EtOH) administered to rodents, by monitoring alcohol metabolic enzymes, oxidative stress signals, and inflammatory mediators. The effect of tyrosine-alanine (YA) peptide, a main component of OH, on EtOH metabolism was also identified. In vitro experiments showed that OH pretreatment inhibited EtOH-induced cell death, oxidative stress, and inflammation in liver cells and macrophages. In vivo experiments showed that OH and YA pre-administration increased alcohol dehydrogenase, aldehyde dehydrogenase, and catalase activity in EtOH binge treatment. In addition, OH pre-administration alleviated CYP2E1 activity, ROS production, apoptotic signals, and inflammatory mediators in liver tissues. These results showed that OH and YA enhanced EtOH metabolism and had a protective effect against acute alcohol liver damage. Our findings offer new insights into a single high dose of EtOH drinking and suggest that OH and YA could be used as potential marine functional foods to prevent acute alcohol-induced liver damage.
Collapse
Affiliation(s)
- Adrian S. Siregar
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
| | - Marie Merci Nyiramana
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
| | - Eun-Jin Kim
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
| | - Eui-Jung Shin
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
| | - Min Seok Woo
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
| | - Jin-Mok Kim
- Department of Clinical Laboratory Science, Masan University, Changwon 2640, Korea;
| | - Jung Hwan Kim
- Department of Premedicine, College of Medicine, Gyeongsang National University, Jinju 52727, Korea;
| | - Dong Kun Lee
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
| | - Jong Ryeal Hahm
- Department of Internal Medicine, Hospital and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea;
| | - Hyun Joon Kim
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
- Department of Anatomy and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Chang-Woon Kim
- Department of Obstetrics and Gynecology, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Korea;
| | - Nam-Gil Kim
- Department of Marine Biology and Aquaculture and Institute of Marine Industry, Gyeongsang National University, Tongyeong 53064, Korea;
| | - Si-Hyang Park
- Sunmarin Biotech, Jinju Bioindustry Foundation, Jinju 52839, Korea;
| | - Yeung Joon Choi
- Ocean-Pep, Jinju Bioindustry Foundation, Jinju 52839, Korea;
| | - Sang Soo Kang
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
- Department of Anatomy and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Seong-Geun Hong
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
| | - Jaehee Han
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
| | - Dawon Kang
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
- Correspondence:
| |
Collapse
|
19
|
Xu H, He K, Li Y, Tao Y, Xu C, Hu Z, Wang T, Zhang C. Cytoprotective Effects Evaluation of a Novel Danshensu Derivative DEX-018 against Oxidative Stress Injury in HUVECs. Biol Pharm Bull 2020; 43:801-809. [PMID: 32132313 DOI: 10.1248/bpb.b19-00878] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ischemic heart disease (IHD) is one of the most common cardiovascular diseases with high morbidity and mortality. Danshensu (DSS) is widely used in the treatment of coronary heart disease. In this study, the carboxy group of DSS was esterified with edaravone to synthesize the novel DSS derivative DEX-018 to achieve a synergistic protective effect and overcome the structural deficiency of DSS. The pharmacological effect of DEX-018 against tert-butyl hydrogen peroxide (t-BHP) induced oxidative damage in human umbilical vein endothelial cells (HUVECs) was evaluated. The results demonstrated that pretreatment with DEX-018 significantly increased cell viability and superoxide dismutase (SOD) activity and decreased the lactate dehydrogenase (LDH) leakage rate, malondialdehyde (MDA) level and intracellular reactive oxygen species (ROS) level. In addition, DEX-018 inhibited cell apoptosis and reversed the expression of apoptosis-related proteins (Bcl-2, Bax, and caspase-3) in HUVECs stimulated by t-BHP. Further study on the mechanism of DEX-018 revealed that the expression of p-Akt and p-extracellular signal-regulated kinase 1/2 (ERK1/2) was increased, which suggested that DEX-018 may protect HUVECs against t-BHP induced oxidative injury via the Akt and ERK1/2 signaling pathways. To further validate the correlation, CCK8 was used to detect cell viability after treatment with DEX-018 plus Akt inhibitor (MK2206) and phosphadylinositol 3-kinase (PI3K) inhibitor (LY294002). Compared with DEX-018 alone, MK2206 or LY294002 significantly decreased cell viability of HUVECs, indicating that the protective effect of DEX-018 against t-BHP induced oxidative injury was significantly weakened. It was further verified that the antioxidant and anti-apoptotic effects of DEX-018 were partly related to the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Honglei Xu
- School of Pharmacy, Second Military Medical University
| | - Kun He
- School of Pharmacy, Second Military Medical University
| | - Yi Li
- Shanghai University of Traditional Chinese Medicine
| | - Yulong Tao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University
| | - Chunfang Xu
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University
| | - Zhenlin Hu
- School of Pharmacy, Second Military Medical University
| | | | - Chuan Zhang
- School of Pharmacy, Second Military Medical University.,School of Medicine, Shanghai University
| |
Collapse
|
20
|
Srinivasan MP, Bhopale KK, Amer SM, Wan J, Kaphalia L, Ansari GS, Kaphalia BS. Linking Dysregulated AMPK Signaling and ER Stress in Ethanol-Induced Liver Injury in Hepatic Alcohol Dehydrogenase Deficient Deer Mice. Biomolecules 2019; 9:biom9100560. [PMID: 31581705 PMCID: PMC6843321 DOI: 10.3390/biom9100560] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/23/2019] [Accepted: 09/29/2019] [Indexed: 12/12/2022] Open
Abstract
Ethanol (EtOH) metabolism itself can be a predisposing factor for initiation of alcoholic liver disease (ALD). Therefore, a dose dependent study to evaluate liver injury was conducted in hepatic alcohol dehydrogenase (ADH) deficient (ADH−) and ADH normal (ADH+) deer mice fed 1%, 2% or 3.5% EtOH in the liquid diet daily for 2 months. Blood alcohol concentration (BAC), liver injury marker (alanine amino transferase (ALT)), hepatic lipids and cytochrome P450 2E1 (CYP2E1) activity were measured. Liver histology, endoplasmic reticulum (ER) stress, AMP-activated protein kinase (AMPK) signaling and cell death proteins were evaluated. Significantly increased BAC, plasma ALT, hepatic lipids and steatosis were found only in ADH− deer mice fed 3.5% EtOH. Further, a significant ER stress and increased un-spliced X-box binding protein 1 were evident only in ADH− deer mice fed 3.5% EtOH. Both strains fed 3.5% EtOH showed deactivation of AMPK, but increased acetyl Co-A carboxylase 1 and decreased carnitine palmitoyltransferase 1A favoring lipogenesis were found only in ADH− deer mice fed 3.5% EtOH. Therefore, irrespective of CYP2E1 overexpression; EtOH dose and hepatic ADH deficiency contribute to EtOH-induced steatosis and liver injury, suggesting a linkage between ER stress, dysregulated hepatic lipid metabolism and AMPK signaling.
Collapse
Affiliation(s)
- Mukund P Srinivasan
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kamlesh K Bhopale
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Samir M Amer
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Forensic Medicine and Clinical Toxicology, Tanta University, Tanta 31512, Egypt
| | - Jie Wan
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Lata Kaphalia
- Division of Pulmonary, Critical Care Medicine, Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ghulam S Ansari
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Bhupendra S Kaphalia
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|