1
|
Yang J, Yang Y, Tan X, Du H, Zhou Z, Chen L, Tian X, Zheng G, Hu J, Zhang C, Qiu Z. Unlocking the potential of the ACE2/Ang-(1-7)/Mas Axis in liver diseases: From molecular mechanisms to translational applications. Diabetes Obes Metab 2025. [PMID: 40344459 DOI: 10.1111/dom.16435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
Over the past two decades, the identification of new functions within the renin-angiotensin system (RAS) has extended beyond its traditional roles, with the emergence of the angiotensin-converting enzyme 2 (ACE2)/Ang-(1-7)/Mas axis being particularly significant. This axis is hypothesized to balance or modulate the effects of the traditional ACE/Ang II/AT1 axis in various physiological and pathological contexts. ACE2, a membrane-bound carboxypeptidase and an ancient homologue of ACE converts Angiotensin II (Ang II) into Angiotensin 1-7 (Ang-(1-7)). The Mas receptor is a G-protein-coupled receptor that specifically binds Ang-(1-7). Recent research has increasingly focused on the local expression of RAS in different tissues. Ang-(1-7) produces a variety of biological effects by binding to the Mas receptor, including anti-inflammatory, antioxidant, anti-apoptotic and anti-fibrotic actions, thereby influencing a range of mechanisms in the heart, kidneys, brain and other tissues. Preclinical animal model studies indicate that manipulating the protective RAS can significantly alter the progression of multiple liver diseases. Hepatic overexpression of ACE2 or administration of Ang-(1-7) and its analogues has been shown to be therapeutically effective against drug-induced liver injury, metabolic-associated fatty liver disease, liver fibrosis and hepatocellular carcinoma progression. These effects are achieved through various pathways, including the regulation of lipid metabolism, inhibition of epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM) production, as well as suppression of aerobic glycolysis. In current clinical trials, while recombinant human ACE2 (Rh-ACE2) has demonstrated safety and good tolerance in most studies, research on the relevance of activating the ACE2/Ang-(1-7) axis in the mechanisms and evolution of human diseases remains in its early stages. Therefore, further elucidation of the complex interactions between the classical and counter-regulatory RAS axes in clinical settings is crucial. This review will summarize the roles of selective activation of the ACE2/Ang-(1-7)/Mas axis, with a focus on its mechanisms in the treatment of liver diseases. Additionally, we will discuss the safety concerns regarding selective activation of the ACE2/Ang-(1-7)/Mas axis in clinical applications and the challenges of tissue-specific activation of this axis, providing effective therapeutic strategies for targeted activation of the hepatic ACE2/Ang-(1-7)/Mas axis in clinical practice.
Collapse
Affiliation(s)
- Jun Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Yuan Yang
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiangyun Tan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Hongzhi Du
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Zhongshi Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Liang Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Xianxiang Tian
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Guohua Zheng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Junjie Hu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Cong Zhang
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Zhenpeng Qiu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Hubei Shizhen Laboratory, Wuhan, People's Republic of China
| |
Collapse
|
2
|
Awashra A, Nouri A, Hamdan A, Said H, Rajab I, Hussein A, Abu Rmilah A. Metabolic Dysfunction-Associated Steatotic Liver Disease as a Cardiovascular Risk Factor: Focus on Atrial Fibrillation. Cardiol Rev 2025:00045415-990000000-00473. [PMID: 40262019 DOI: 10.1097/crd.0000000000000932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as nonalcoholic fatty liver disease, is increasingly recognized as a multisystem disorder with significant cardiovascular implications, particularly in its association with atrial fibrillation (AF). As the most common sustained cardiac arrhythmia, AF contributes to substantial morbidity and mortality, making it essential to explore its links with MASLD. The relationship between these conditions is underpinned by shared pathophysiological mechanisms, including systemic inflammation, insulin resistance, oxidative stress, and activation of the renin-angiotensin-aldosterone system. These processes drive atrial remodeling and electrical instability, predisposing individuals with MASLD to AF. Epidemiological studies further support this connection, showing an independent association between MASLD and an increased risk of AF, particularly in those with metabolic comorbidities such as obesity and type 2 diabetes. Beyond increasing AF susceptibility, MASLD may also influence disease progression and response to treatment, affecting anticoagulation safety, rhythm-control strategies, and the success of catheter ablation. Given these clinical implications, therapies targeting metabolic dysfunction-such as statins, renin-angiotensin-aldosterone system inhibitors, and structured lifestyle modifications-may offer dual benefits in mitigating both MASLD and AF risk. However, significant knowledge gaps remain regarding the causal direction of this association, the impact of MASLD severity on AF burden, and the most effective management strategies for patients with both conditions. Future research should prioritize longitudinal studies, mechanistic investigations, and randomized controlled trials to deepen our understanding of this relationship, ultimately guiding more personalized and integrated treatment approaches. Incorporating MASLD screening into cardiovascular risk assessment may enhance early detection and improve outcomes for at-risk populations.
Collapse
Affiliation(s)
- Ameer Awashra
- From the Department of Medicine, Mayo Clinic, Rochester, MN
| | - Ahmad Nouri
- From the Department of Medicine, Mayo Clinic, Rochester, MN
- Department of Medicine, Qatar University, Doha, Qatar
| | - Ahmad Hamdan
- From the Department of Medicine, Mayo Clinic, Rochester, MN
| | - Hanin Said
- From the Department of Medicine, Mayo Clinic, Rochester, MN
| | - Islam Rajab
- Department of Internal Medicine, St. Joseph's University Medical Center, Paterson, NJ
| | - Abdallah Hussein
- Internal Medicine Department, Virtua Our Lady of Lourdes Hospital, Camden, NJ
| | - Anan Abu Rmilah
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
3
|
Gao Y, Chen Q, Yang S, Cao J, Li F, Li R, Wu Z, Wang Y, Yuan L. Indole alleviates nonalcoholic fatty liver disease in an ACE2-dependent manner. FASEB J 2024; 38:e70061. [PMID: 39305120 DOI: 10.1096/fj.202401172rr] [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: 05/25/2024] [Revised: 08/27/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024]
Abstract
Indole is a microbial metabolite produced by the gut microbiota through the degradation of dietary tryptophan, known for its well-established anti-inflammatory and antioxidant properties. In this study, we collected fecal samples from mice fed a high-fat diet (HFD) and those on a standard diet (SD), then conducted 16S rRNA sequencing to analyze their gut microbiota. The analysis revealed distinct differences in the dominant bacterial species between the two groups, with a significant decrease in indole-producing probiotics in the HFD mice compared to the SD group. Then we administered oral indole treatment to male C57BL/6J mice with HFD-induced NAFLD and observed a significant improvement in hepatic steatosis and inflammation. Notably, indole alleviated the HFD-induced decline in serum Angiotensin-(1-7) [Ang-(1-7)] levels and Angiotensin-Converting Enzyme 2 (ACE2) expression. To further investigate the role of indole and ACE2 in NAFLD, we conducted experiments using ACE2 knockout (ACE2KO) mice that were also induced with HFD-induced NAFLD and treated with indole. Interestingly, the protective effects of indole were compromised in the absence of ACE2. In HepG2 cells, indole similarly stimulated ACE2 expression and, in an ACE2-dependent manner, reduced ROS generation, maintained mitochondrial membrane potential stability, and increased SIRT3 expression. In summary, our results highlight the formation of a biologically active gut-liver axis between the gut microbiota and the liver through the tryptophan metabolite indole, which mitigates NAFLD in an ACE2-dependent manner. Elevating dietary tryptophan and increasing indole levels may represent an effective approach for preventing and treating NAFLD.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songtao Yang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Cao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangyu Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuoying Wu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Wang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Alvarado-Ojeda ZA, Trejo-Moreno C, Ferat-Osorio E, Méndez-Martínez M, Fragoso G, Rosas-Salgado G. Role of Angiotensin II in Non-Alcoholic Steatosis Development. Arch Med Res 2024; 55:102986. [PMID: 38492325 DOI: 10.1016/j.arcmed.2024.102986] [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: 08/30/2023] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Fatty liver is a multifactorial disease characterized by excessive accumulation of lipids in hepatocytes (steatosis), insulin resistance, oxidative stress, and inflammation. This disease has a major public health impact because it is the first stage of a chronic and degenerative process in the liver that can lead to steatohepatitis, cirrhosis, and liver cancer. Although this disease is mainly diagnosed in patients with obesity, type 2 diabetes mellitus, and dyslipidemia, recent evidence indicates that vasoactive hormones such as angiotensin II (ANGII) not only promote endothelial dysfunction (ED) and hypertension, but also cause fatty liver, increase adipose tissue, and develop a pro-steatotic environment characterized by a low-grade systemic pro-inflammatory and pro-oxidant state, with elevated blood lipid levels. The role of ANGII in lipid accumulation has been little studied, so this review aims to summarize existing reports on the possible mechanism of action of ANGII in inducing lipid accumulation in hepatocytes.
Collapse
Affiliation(s)
| | - Celeste Trejo-Moreno
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca Morelos, Mexico
| | - Eduardo Ferat-Osorio
- División de Investigación en Salud, Unidad de Investigación en Epidemiología Clínica, Hospital de Especialidades, Dr. Bernardo Sepúlveda Gutiérrez, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Marisol Méndez-Martínez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Gladis Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gabriela Rosas-Salgado
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca Morelos, Mexico.
| |
Collapse
|
5
|
Zhou L, Chen S, Wei Y, Sun Y, Yang Y, Lin B, Li Y, Wang C. Glycyrrhizic acid restores the downregulated hepatic ACE2 signaling in the attenuation of mouse steatohepatitis. Eur J Pharmacol 2024; 967:176365. [PMID: 38316247 DOI: 10.1016/j.ejphar.2024.176365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Glycyrrhizic acid (GA), one of the major active components derived from licorice root, exerts liver-protecting activity. Its molecular mechanisms of action, however, remain not completely understood. The angiotensin (Ang) converting enzyme (ACE) 2/Ang-(1-7)/Mas axis, regulated by ACE2 through converting Ang II into Ang-(1-7) to activate Mas receptor, counteracts the pro-inflammatory and pro-steatotic effects of the ACE/Ang II/Ang II receptor type 1 (AT1) axis. Here, it was found that pretreatment with GA suppressed LPS/D-galactosamine-induced serum hyperactivities of alanine aminotransferase and aspartate aminotransferase, hepatomegaly, pathological changes, and over-accumulation of triglycerides and fatty droplets in the liver of mice. GA also diminished LPS/free fatty acid-induced inflammation and steatosis in cultured hepatocytes. Mechanistically, GA restored hepatic protein hypoexpression of ACE2 and Mas receptor, and the decrease in hepatic Ang-(1-7) content. Hepatic overexpression of angiotensin II and AT1 was also suppressed. However, GA did not alter hepatic protein expression of renin and ACE. In addition, GA inhibited hepatic protein over-phosphorylation of the p38 mitogen-activated protein kinase, c-Jun N-terminal kinase, extracellular signal-regulated kinase, and nuclear factor κB at Ser536. Hepatic overexpression of tumor necrosis factor α, interleukin 6, interleukin 1β, sterol regulatory element-binding protein 1c, and fatty acid synthase was also inhibited. GA-elicited recovery of ACE2 and Mas protein hypoexpression was further confirmed in the hepatocyte. Thus, the present results demonstrate that GA restores the downregulated hepatic ACE2-mediated anti-inflammatory and anti-steatotic signaling in the amelioration of steatohepatitis. We suggest that GA may protect the liver from injury by regulating the hepatic ACE2-mediated signaling.
Collapse
Affiliation(s)
- Longyue Zhou
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Shankang Chen
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yuanyi Wei
- Department of Pharmacy, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Yihui Sun
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Yifan Yang
- Endocrinology and Metabolism Group, Sydney Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, NSW, 2000, Australia.
| | - Bingqi Lin
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yuhao Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Endocrinology and Metabolism Group, Sydney Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, NSW, 2000, Australia.
| | - Chunxia Wang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
6
|
Zuo Q, Park NH, Lee JK, Santaliz-Casiano A, Madak-Erdogan Z. Navigating nonalcoholic fatty liver disease (NAFLD): Exploring the roles of estrogens, pharmacological and medical interventions, and life style. Steroids 2024; 203:109330. [PMID: 37923152 DOI: 10.1016/j.steroids.2023.109330] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
The pursuit of studying this subject is driven by the urgency to address the increasing global prevalence of Non-Alcoholic Fatty Liver Disease (NAFLD) and its profound health implications. NAFLD represents a significant public health concern due to its association with metabolic disorders, cardiovascular complications, and the potential progression to more severe conditions like non-alcoholic steatohepatitis (NASH), fibrosis, and cirrhosis. Liver estrogen signaling is important for maintaining liver function, and loss of estrogens increases the likelihood of NAFLD in postmenopausal women. Understanding the multifaceted mechanisms underlying NAFLD pathogenesis, its varied treatment strategies, and their effectiveness is crucial for devising comprehensive and targeted interventions. By unraveling the intricate interplay between genetics, lifestyle, hormonal regulation, and gut microbiota, we can unlock insights into risk stratification, early detection, and personalized therapeutic approaches. Furthermore, investigating the emerging pharmaceutical interventions and dietary modifications offers the potential to revolutionize disease management. This review reinforces the role of collaboration in refining NAFLD comprehension, unveiling novel therapeutic pathways, and ultimately improving patient outcomes for this intricate hepatic condition.
Collapse
Affiliation(s)
- Qianying Zuo
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Nicole Hwajin Park
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Jenna Kathryn Lee
- Department of Neuroscience, Northwestern University, Evanston, IL 60208, USA
| | - Ashlie Santaliz-Casiano
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Zeynep Madak-Erdogan
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
7
|
Chen L, Ye X, Yang L, Zhao J, You J, Feng Y. Linking fatty liver diseases to hepatocellular carcinoma by hepatic stellate cells. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:25-35. [PMID: 39036388 PMCID: PMC11256631 DOI: 10.1016/j.jncc.2024.01.002] [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: 11/30/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 07/23/2024] Open
Abstract
Hepatic stellate cells (HSCs), a distinct category of non-parenchymal cells in the liver, are critical for liver homeostasis. In healthy livers, HSCs remain non-proliferative and quiescent. However, under conditions of acute or chronic liver damage, HSCs are activated and participate in the progression and regulation of liver diseases such as liver fibrosis, cirrhosis, and liver cancer. Fatty liver diseases (FLD), including nonalcoholic (NAFLD) and alcohol-related (ALD), are common chronic inflammatory conditions of the liver. These diseases, often resulting from multiple metabolic disorders, can progress through a sequence of inflammation, fibrosis, and ultimately, cancer. In this review, we focused on the activation and regulatory mechanism of HSCs in the context of FLD. We summarized the molecular pathways of activated HSCs (aHSCs) in mediating FLD and their role in promoting liver tumor development from the perspectives of cell proliferation, invasion, metastasis, angiogenesis, immunosuppression, and chemo-resistance. We aimed to offer an in-depth discussion on the reciprocal regulatory interactions between FLD and HSC activation, providing new insights for researchers in this field.
Collapse
Affiliation(s)
- Liang'en Chen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Xiangshi Ye
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Lixian Yang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital (Hangzhou Medical College), Hangzhou, China
| | - Jiangsha Zhao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jia You
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Yuxiong Feng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Zou L, Yu X, Xiong J, Chen C, Xiao G. Partial Replacement of NaCl with KCl in Cooked Meat Could Reduce the Liver Damage Through Renin-Angiotensin System in Mice. Mol Nutr Food Res 2024; 68:e2200783. [PMID: 38308101 DOI: 10.1002/mnfr.202200783] [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: 11/11/2022] [Revised: 05/29/2023] [Indexed: 02/04/2024]
Abstract
SCOPE Dietary salt (sodium chloride, NaCl) is necessary for processed meat products, but intake of a high-sodium diet carries serious health risks. Considerable studies indicate that the partial substitution of NaCl with potassium chloride (KCl) can produce sodium-reduced cooked meat. However, most studies of sodium-reduced cooked meat focus on the production process in vitro, and the effect of cooked meat on health has not been well clarified in vivo. METHODS AND RESULTS This study finds that compared to the high-sodium group (HS), serum renin, angiotensin-converting enzyme (ACE), angiotensin (Ang) II, and the levels of some indicators of dyslipidemia are decreased in the reduced salt by partial substitution of NaCl with KCl group (RS + K). Furthermore, RS + K increases the antioxidation abilities, inhibits the renin-angiotensin system (RAS) through ACE/Ang II/Ang II type 1 receptor axis pathway, reduces synthesis of triglyceride and cholesterol and protein expressions of inflammatory factors interleukin-17A and nuclear factor-kappa B in the liver. CONCLUSION Partial substitution of NaCl with KCl in cooked meat can be a feasible approach for improving the health benefits and developing novel functional meat products for nutritional health interventions.
Collapse
Affiliation(s)
- Lifang Zou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, 230009, China
| | - Xia Yu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, 230009, China
| | - Jiahao Xiong
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, 230009, China
| | - Conggui Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, 230009, China
- Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei, 230009, China
| | - Guiran Xiao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, 230009, China
| |
Collapse
|
9
|
Aziel Alvarado-Ojeda Z, Zamilpa A, Costet-Mejia A, Méndez-Martínez M, Trejo-Moreno C, Jiménez-Ferrer JE, Salazar-Martínez AM, Cruz-Muñoz ME, Fragoso G, Rosas-Salgado G. Hydroalcoholic extract from Sechium edule (Jacq.) S.w. root reverses oleic acid-induced steatosis and insulin resistance in vitro. Heliyon 2024; 10:e24567. [PMID: 38312619 PMCID: PMC10835324 DOI: 10.1016/j.heliyon.2024.e24567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/17/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Steatosis is characterized by fat accumulation and insulin resistance (IR) in hepatocytes, which triggers a pro-oxidant, pro-inflammatory environment that may eventually lead to cirrhosis or liver carcinoma. This work was aimed to assess the effect of Sechium edule root hydroalcoholic extract (rSe-HA) (rich in cinnamic and coumaric acid, among other phenolic compounds) on triglyceride esterification, lipid degradation, AMPK expression, and the phosphorylation of insulin receptor in a Ser312 residue, as well as on the redox status, malondialdehyde (MDA) production, and the production of proinflammatory cytokines in an in vitro model of steatosis induced by oleic acid, to help develop a phytomedicine that could reverse this pathology. rSe-HA reduced triglyceride levels in hepatocyte lysates, increased lipolysis by activating AMPK at Thr172, and improved the redox status, as evidenced by the concentration of glycerol and formazan, respectively. It also prevented insulin resistance (IR), as measured by glucose consumption and the phosphorylation of the insulin receptor at Ser312. It also prevented TNFα and IL6 production and decreased the levels of MDA and nitric oxide (ON). Our results indicate that rSe-HA reversed steatosis and controlled the proinflammatory and prooxidant environment in oleic acid-induced dysfunctional HepG2 hepatocytes, supporting its potential use to control this disorder.
Collapse
Affiliation(s)
- Zimri Aziel Alvarado-Ojeda
- Facultad de Medicina, Universidad Autónoma Del Estado de Morelos, Leñeros S/N, Cuernavaca, Morelos, 62350, Mexico
| | - Alejandro Zamilpa
- Laboratorio de Farmacología, Centro de Investigaciones Biomédicas Del Sur, Instituto Mexicano Del Seguro Social, Xochitepec, Morelos, 62790, Mexico
| | - Alejandro Costet-Mejia
- Laboratorio de Farmacología, Centro de Investigaciones Biomédicas Del Sur, Instituto Mexicano Del Seguro Social, Xochitepec, Morelos, 62790, Mexico
| | - Marisol Méndez-Martínez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana-Xochimilco, 04960, Mexico City, Mexico
| | - Celeste Trejo-Moreno
- Facultad de Medicina, Universidad Autónoma Del Estado de Morelos, Leñeros S/N, Cuernavaca, Morelos, 62350, Mexico
| | - Jesús Enrique Jiménez-Ferrer
- Laboratorio de Farmacología, Centro de Investigaciones Biomédicas Del Sur, Instituto Mexicano Del Seguro Social, Xochitepec, Morelos, 62790, Mexico
| | - Ana Maria Salazar-Martínez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Mario Ernesto Cruz-Muñoz
- Facultad de Medicina, Universidad Autónoma Del Estado de Morelos, Leñeros S/N, Cuernavaca, Morelos, 62350, Mexico
| | - Gladis Fragoso
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Gabriela Rosas-Salgado
- Facultad de Medicina, Universidad Autónoma Del Estado de Morelos, Leñeros S/N, Cuernavaca, Morelos, 62350, Mexico
| |
Collapse
|
10
|
Driessen S, Francque SM, Anker SD, Castro Cabezas M, Grobbee DE, Tushuizen ME, Holleboom AG. Metabolic dysfunction-associated steatotic liver disease and the heart. Hepatology 2023:01515467-990000000-00699. [PMID: 38147315 DOI: 10.1097/hep.0000000000000735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/13/2023] [Indexed: 12/27/2023]
Abstract
The prevalence and severity of metabolic dysfunction-associated steatotic liver disease (MASLD) are increasing. Physicians who treat patients with MASLD may acknowledge the strong coincidence with cardiometabolic disease, including atherosclerotic cardiovascular disease (asCVD). This raises questions on co-occurrence, causality, and the need for screening and multidisciplinary care for MASLD in patients with asCVD, and vice versa. Here, we review the interrelations of MASLD and heart disease and formulate answers to these matters. Epidemiological studies scoring proxies for atherosclerosis and actual cardiovascular events indicate increased atherosclerosis in patients with MASLD, yet no increased risk of asCVD mortality. MASLD and asCVD share common drivers: obesity, insulin resistance and type 2 diabetes mellitus (T2DM), smoking, hypertension, and sleep apnea syndrome. In addition, Mendelian randomization studies support that MASLD may cause atherosclerosis through mixed hyperlipidemia, while such evidence is lacking for liver-derived procoagulant factors. In the more advanced fibrotic stages, MASLD may contribute to heart failure with preserved ejection fraction by reduced filling of the right ventricle, which may induce fatigue upon exertion, often mentioned by patients with MASLD. Some evidence points to an association between MASLD and cardiac arrhythmias. Regarding treatment and given the strong co-occurrence of MASLD and asCVD, pharmacotherapy in development for advanced stages of MASLD would ideally also reduce cardiovascular events, as has been demonstrated for T2DM treatments. Given the common drivers, potential causal factors and especially given the increased rate of cardiovascular events, comprehensive cardiometabolic risk management is warranted in patients with MASLD, preferably in a multidisciplinary approach.
Collapse
Affiliation(s)
- Stan Driessen
- Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Sven M Francque
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, Antwerp, Belgium
| | - Stefan D Anker
- Department of Cardiology (CVK) of German Heart Center Charité, Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Manuel Castro Cabezas
- Julius Clinical, Zeist, The Netherlands
- Department of Internal Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, The Netherlands
- Department of Internal Medicine and Endocrinology, Erasmus MC, Rotterdam, The Netherlands
| | - Diederick E Grobbee
- Julius Clinical, Zeist, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maarten E Tushuizen
- Department of Gastroenterology and Hepatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Adriaan G Holleboom
- Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Kazura W, Michalczyk K, Skrzep-Poloczek B, Chełmecka E, Zalejska-Fiolka J, Michalski M, Kukla M, Jochem J, Rutkowski J, Stygar D. Liver Oxidative Status, Serum Lipids Levels after Bariatric Surgery and High-Fat, High-Sugar Diet in Animal Model of Induced Obesity. Int J Mol Sci 2023; 24:16535. [PMID: 38003721 PMCID: PMC10671458 DOI: 10.3390/ijms242216535] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Nutritional status is a major determinant of hepatocyte injuries associated with changed metabolism and oxidative stress. This study aimed to determine the relations between oxidative stress, bariatric surgery, and a high-fat/high-sugar (HFS) diet in a diet-induced obesity rat model. Male rats were maintained on a control diet (CD) or high-fat/high-sugar diet (HFS) inducing obesity. After 8 weeks, the animals underwent SHAM (n = 14) or DJOS (n = 14) surgery and the diet was either changed or unchanged. Eight weeks after the surgeries, the activity of superoxide dismutase isoforms (total SOD, MnSOD, and CuZnSOD), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), and lutathione S-transferase, as well as the thiol groups (-SH) concentration, total antioxidant capacity (TAC), total oxidative stress (TOS) levels, and malondialdehyde (MDA) concentration liver tissue were assessed. The total cholesterol, triglycerides (TG), and high-density lipoprotein (HDL) concentrations were measured in the serum. The total SOD and GPX activities were higher in the SHAM-operated rats than in the DJOS-operated rats. The MnSOD activity was higher in the HFS/HFS than the CD/CD groups. Higher CuZnSOD, GST, GR activities, -SH, and MDA concentrations in the liver, and the triglyceride and cholesterol concentrations in the serum were observed in the SHAM-operated rats than in the DJOS-operated rats. The CAT activity was significantly higher in the HFS-fed rats. Lower TAC and higher TOS values were observed in the SHAM-operated rats. Unhealthy habits after bariatric surgery may be responsible for treatment failure and establishing an obesity condition with increased oxidative stress.
Collapse
Affiliation(s)
- Wojciech Kazura
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana Street, 41-808 Zabrze, Poland
| | - Katarzyna Michalczyk
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana Street, 41-808 Zabrze, Poland
| | - Bronisława Skrzep-Poloczek
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana Street, 41-808 Zabrze, Poland
| | - Elżbieta Chełmecka
- Department of Medical Statistics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 31 Ostrogórska Street, 41-200 Sosnowiec, Poland
| | - Jolanta Zalejska-Fiolka
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana Street, 41-808 Zabrze, Poland
| | - Marek Michalski
- Department of Histology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana Street, 41-808 Zabrze, Poland
| | - Michał Kukla
- Department of Internal Medicine and Geriatrics, Faculty of Medicine, Jagellonian University Medical College, 31-688 Kraków, Poland
- Department of Endoscopy, University Hospital, 30-688 Kraków, Poland
| | - Jerzy Jochem
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana Street, 41-808 Zabrze, Poland
| | - Jakub Rutkowski
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana Street, 41-808 Zabrze, Poland
| | - Dominika Stygar
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana Street, 41-808 Zabrze, Poland
- SLU University Animal Hospital, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| |
Collapse
|
12
|
Trejo-Moreno C, Alvarado-Ojeda ZA, Méndez-Martínez M, Cruz-Muñoz ME, Castro-Martínez G, Arrellín-Rosas G, Zamilpa A, Jimenez-Ferrer JE, Baez Reyes JC, Fragoso G, Salgado GR. Aqueous Fraction from Cucumis sativus Aerial Parts Attenuates Angiotensin II-Induced Endothelial Dysfunction In Vivo by Activating Akt. Nutrients 2023; 15:4680. [PMID: 37960332 PMCID: PMC10649625 DOI: 10.3390/nu15214680] [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: 10/12/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Endothelial dysfunction (ED) is a marker of vascular damage and a precursor of cardiovascular diseases such as hypertension, which involve inflammation and organ damage. Nitric oxide (NO), produced by eNOS, which is induced by pAKT, plays a crucial role in the function of a healthy endothelium. METHODS A combination of subfractions SF1 and SF3 (C4) of the aqueous fraction from Cucumis sativus (Cs-Aq) was evaluated to control endothelial dysfunction in vivo and on HMEC-1 cells to assess the involvement of pAkt in vitro. C57BL/6J mice were injected daily with angiotensin II (Ang-II) for 10 weeks. Once hypertension was established, either Cs-AqC4 or losartan was orally administered along with Ang-II for a further 10 weeks. Blood pressure (BP) was measured at weeks 0, 5, 10, 15, and 20. In addition, serum creatinine, inflammatory status (in the kidney), tissue damage, and vascular remodeling (in the liver and aorta) were evaluated. Cs-AqC4 was also tested in vitro on HMEC-1 cells stimulated by Ang-II to assess the involvement of Akt phosphorylation. RESULTS Cs-AqC4 decreased systolic and diastolic BP, reversed vascular remodeling, decreased IL-1β and TGF-β, increased IL-10, and decreased kidney and liver damage. In HMEC-1 cells, AKT phosphorylation and NO production were increased. CONCLUSIONS Cs-AqC4 controlled inflammation and vascular remodeling, alleviating hypertension; it also improved tissue damage associated with ED, probably via Akt activation.
Collapse
Affiliation(s)
- Celeste Trejo-Moreno
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Morelos, Mexico; (C.T.-M.); (Z.A.A.-O.); (M.E.C.-M.); (G.A.-R.)
| | - Zimri Aziel Alvarado-Ojeda
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Morelos, Mexico; (C.T.-M.); (Z.A.A.-O.); (M.E.C.-M.); (G.A.-R.)
| | - Marisol Méndez-Martínez
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Ciudad de México 04960, Mexico;
| | - Mario Ernesto Cruz-Muñoz
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Morelos, Mexico; (C.T.-M.); (Z.A.A.-O.); (M.E.C.-M.); (G.A.-R.)
| | - Gabriela Castro-Martínez
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 04960, Mexico;
| | - Gerardo Arrellín-Rosas
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Morelos, Mexico; (C.T.-M.); (Z.A.A.-O.); (M.E.C.-M.); (G.A.-R.)
- Facultad de Ciencias de la Salud, Universidad Panamericana, Ciudad de México 03920, Mexico
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec 62790, Morelos, Mexico; (A.Z.); (J.E.J.-F.)
| | - Jesús Enrique Jimenez-Ferrer
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec 62790, Morelos, Mexico; (A.Z.); (J.E.J.-F.)
| | - Juan Carlos Baez Reyes
- Escuela Nacional Preparatoria No. 1, Universidad Nacional Autónoma de México, Ciudad de México 16030, Mexico;
| | - Gladis Fragoso
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Gabriela Rosas Salgado
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Morelos, Mexico; (C.T.-M.); (Z.A.A.-O.); (M.E.C.-M.); (G.A.-R.)
| |
Collapse
|
13
|
Papaefthymiou A, Doulberis M, Karafyllidou K, Chatzimichael E, Deretzi G, Exadaktylos AK, Sampsonas F, Gelasakis A, Papamichos SI, Kotronis G, Gialamprinou D, Vardaka E, Polyzos SA, Kountouras J. Effect of spironolactone on pharmacological treatment of nonalcoholic fatty liver disease. Minerva Endocrinol (Torino) 2023; 48:346-359. [PMID: 34669319 DOI: 10.23736/s2724-6507.21.03564-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) was recently renamed to metabolic (dysfunction)-associated fatty liver disease (MAFLD) to better characterize its pathogenic origin. NAFLD represents, at least in western societies, a potential epidemic with raising prevalence. Its multifactorial pathogenesis is partially unraveled and till now there is no approved pharmacotherapy for NAFLD. A plethora of various choices are investigated in clinical trials, targeting an arsenal of different pathways and molecules. Since the mineralocorticoid receptor (MR) and renin-angiotensin-aldosterone system (RAAS) appear to be implicated in NAFLD, within this concise review, we focus on a rather classical and inexpensive pharmacological agent, spironolactone. We present the current lines of evidence of MR and RAAS-related preclinical models and human trials reporting an association with NAFLD. In conclusion, evidence about spironolactone of RAAS is commented, as potential future pharmacological management of NAFLD.
Collapse
Affiliation(s)
- Apostolis Papaefthymiou
- Department of Gastroenterology, University Hospital of Larisa, Larisa, Greece -
- School of Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece -
- School of Medicine, First Laboratory of Pharmacology, Aristotle University of Thessaloniki, Thessaloniki, Greece -
| | - Michael Doulberis
- School of Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
- School of Medicine, First Laboratory of Pharmacology, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Department of Emergency Medicine, University Hospital Inselspital of Bern, Bern, Switzerland
- Division of Gastroenterology and Hepatology, Department of Medicine, Medical University, Kantonsspital Aarau, Aarau, Switzerland
| | - Kyriaki Karafyllidou
- Department of Pediatrics, University Children's Hospital of Zurich, Zurich, Switzerland
| | - Eleftherios Chatzimichael
- Department of Psychiatry, Psychotherapy and Psychosomatics, Center for Integrative Psychiatry, Psychiatric University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Georgia Deretzi
- Department of Neurology, Papageorgiou General Hospital, Thessaloniki, Greece
| | | | - Fotios Sampsonas
- Department of Respiratory Medicine, University Hospital of Patras, Patras, Greece
| | - Athanasios Gelasakis
- Department of Animal Science, Laboratory of Anatomy and Physiology of Farm Animals, Agricultural University of Athens, Athens, Greece
| | - Spyros I Papamichos
- Blood Transfusion Service Eastern Switzerland, Swiss Red Cross, St. Gallen, Switzerland
| | - Georgios Kotronis
- Department of Internal Medicine, General Hospital Aghios Pavlos of Thessaloniki, Thessaloniki, Greece
| | - Dimitra Gialamprinou
- Second Neonatal Department and NICU, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Elisabeth Vardaka
- School of Health Sciences, Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
| | - Stergios A Polyzos
- School of Medicine, First Laboratory of Pharmacology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Jannis Kountouras
- School of Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
14
|
Fu D, Wu S, Jiang X, You T, Li Y, Xin J, Feng X, Wen J, Huang Y, Hu C. Caveolin-1 alleviates acetaminophen-induced vascular oxidative stress and inflammation in non-alcoholic fatty liver disease. Free Radic Biol Med 2023; 195:245-257. [PMID: 36596386 DOI: 10.1016/j.freeradbiomed.2022.12.095] [Citation(s) in RCA: 4] [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: 11/27/2022] [Revised: 12/24/2022] [Accepted: 12/26/2022] [Indexed: 01/01/2023]
Abstract
Acetaminophen (APAP) is one of the most widely used drugs in the world. The literature shows that excessive or long-term use of APAP can lead to increased cardiovascular dysfunction. An acute increase in angiotensin Ⅱ (Ang Ⅱ) caused by APAP use in fatty liver disease may increase the risk and severity of vascular injury. However, the underlying mechanism remains unclear. Caveolin-1 (CAV1) is a broad-spectrum kinase inhibitor that significantly determines endothelial function. This study aimed to observe the effects of APAP on the vasculature in non-alcoholic fatty liver disease (NAFLD) and to determine whether CAV1 could alleviate vascular oxidative stress and inflammation by targeting Ang Ⅱ or its downstream pathways. In this study, 7-week-old C57BL/6 male mice (18-20 g) were administered APAP by gavage after eight weeks of a high-fat diet. Any resulting vascular oxidative stress and inflammation were assessed. Levels of Ang Ⅱ, CAV1, and other related proteins were measured using ELISA and western blotting. In APAP-treated NAFLD mice, CAV1 expression was downregulated and Ang Ⅱ expression was upregulated compared to normal APAP-treated mice. In vitro, HUVECs were incubated with Ang Ⅱ (300 nM) for 48 h. Overexpression of CAV1 in HUVECs attenuated Ang Ⅱ-induced oxidative stress and inflammation and downregulated the expression of Protein kinase C (PKC) and p-P38/P38. After intervention with CAV1-siRNA, immunofluorescence results showed that the fluorescence intensity of PKC on mitochondria was further increased, and flow cytometry results showed that the mitochondrial membrane potential increased. PKC inhibitors alleviated Ang Ⅱ-induced endothelial injury. In conclusion, our findings confirmed that CAV1 exerts a protective effect against vascular injury by inhibiting oxidative stress and inflammation through the PKC/MAPK pathway. Therefore, restoration of CAV1 may have clinical benefits in reducing APAP-induced vascular damage in NAFLD patients.
Collapse
Affiliation(s)
- Dongdong Fu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Shuai Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Xiangfu Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Tingyu You
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Jiao Xin
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Xiaowen Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Jiagen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Chengmu Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
15
|
Badr AM, Sherif IO, Mahran YF, Attia HA. Role of Renin-Angiotensin System in the Pathogenesis and Progression of Non-alcoholic Fatty Liver. ADVANCES IN BIOCHEMISTRY IN HEALTH AND DISEASE 2023:179-197. [DOI: 10.1007/978-3-031-23621-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
|
16
|
Lee KC, Wu PS, Lin HC. Pathogenesis and treatment of non-alcoholic steatohepatitis and its fibrosis. Clin Mol Hepatol 2023; 29:77-98. [PMID: 36226471 PMCID: PMC9845678 DOI: 10.3350/cmh.2022.0237] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/11/2022] [Indexed: 02/02/2023] Open
Abstract
The initial presentation of non-alcoholic steatohepatitis (NASH) is hepatic steatosis. The dysfunction of lipid metabolism within hepatocytes caused by genetic factors, diet, and insulin resistance causes lipid accumulation. Lipotoxicity, oxidative stress, mitochondrial dysfunction, and endoplasmic reticulum stress would further contribute to hepatocyte injury and death, leading to inflammation and immune dysfunction in the liver. During the healing process, the accumulation of an excessive amount of fibrosis might occur while healing. During the development of NASH and liver fibrosis, the gut-liver axis, adipose-liver axis, and renin-angiotensin system (RAS) may be dysregulated and impaired. Translocation of bacteria or its end-products entering the liver could activate hepatocytes, Kupffer cells, and hepatic stellate cells, exacerbating hepatic steatosis, inflammation, and fibrosis. Bile acids regulate glucose and lipid metabolism through Farnesoid X receptors in the liver and intestine. Increased adipose tissue-derived non-esterified fatty acids would aggravate hepatic steatosis. Increased leptin also plays a role in hepatic fibrogenesis, and decreased adiponectin may contribute to hepatic insulin resistance. Moreover, dysregulation of peroxisome proliferator-activated receptors in the liver, adipose, and muscle tissues may impair lipid metabolism. In addition, the RAS may contribute to hepatic fatty acid metabolism, inflammation, and fibrosis. The treatment includes lifestyle modification, pharmacological therapy, and non-pharmacological therapy. Currently, weight reduction by lifestyle modification or surgery is the most effective therapy. However, vitamin E, pioglitazone, and obeticholic acid have also been suggested. In this review, we will introduce some new clinical trials and experimental therapies for the treatment of NASH and related fibrosis.
Collapse
Affiliation(s)
- Kuei-Chuan Lee
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan,Department of Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan,Corresponding author : Kuei-Chuan Lee Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei 11217, Taiwan Tel: +886 2 2871 2121, Fax: +886 2 2873 9318, E-mail:
| | - Pei-Shan Wu
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan,Department of Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan,Endoscopy Center for Diagnosis and Treatment, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Han-Chieh Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan,Department of Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan,Corresponding author : Kuei-Chuan Lee Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei 11217, Taiwan Tel: +886 2 2871 2121, Fax: +886 2 2873 9318, E-mail:
| |
Collapse
|
17
|
Mahfoz AM, Gawish AY. Insight into the hepatoprotective, hypolipidemic, and antidiabetic impacts of aliskiren in streptozotocin-induced diabetic liver disease in mice. Diabetol Metab Syndr 2022; 14:163. [PMID: 36316746 PMCID: PMC9620647 DOI: 10.1186/s13098-022-00935-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/19/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Diabetic hepatopathy is a serious complication of poorly controlled diabetes mellitus. An efficient antidiabetic drug which keeps normal liver tissues is not available. The renin-angiotensin system has been reported to be involved in both diabetic state and liver function. Aliskiren is a direct renin inhibitor and a recently antihypertensive drug with poly-pharmacological properties. The aim of the current study is to explore the possible hepatoprotective effects and mechanisms of action of aliskiren against streptozotocin (STZ) induced liver toxicity. METHODS Mice were distributed to 3 groups; first: the normal control group, second: the diabetic control group, third: the diabetic group which received aliskiren (25 mg/kg; oral) for 4 weeks. At the end of the treatment period, plasma glucose, insulin, lipid profile, oxidative stress, and liver function tests were evaluated spectrophotometrically. ELISA technique was used to measure the expression levels of TNF-α and adiponectin. Furthermore, a Histopathological examination of liver samples was done. RESULTS It was shown that aliskiren treatment ameliorated the STZ-induced oxidative stress and elevated inflammatory biomarkers, hypercholesterolemia, serum aminotransferases and alkaline phosphatase levels in diabetic mice. In addition, hepatocellular necrosis, and fibrosis were improved by aliskiren treatment. CONCLUSION aliskiren protects against the liver damage caused by STZ-induced diabetes. This can be explained by its ability to block angiotensin-II, and its anti-diabetic, hypocholesterolemic, antioxidant and anti-inflammatory effects. Aliskiren could be a novel therapeutic strategy to prevent liver diseases associated with hypertension and diabetes mellitus.
Collapse
Affiliation(s)
- Amal M Mahfoz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| | - Aya Y Gawish
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| |
Collapse
|
18
|
Yang J, Luo S, Li R, Ju J, Zhang Z, Shen J, Sun M, Fan J, Xia M, Zhu W, Liu Y. Sleep Factors in Relation to Metabolic Dysfunction-Associated Fatty Liver Disease in Middle-Aged and Elderly Chinese. J Clin Endocrinol Metab 2022; 107:2874-2882. [PMID: 35900115 DOI: 10.1210/clinem/dgac428] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Accumulating evidence implies that sleep disturbance is involved in metabolic disorders. OBJECTIVE We comprehensively evaluated the association between various dimensions of sleep behaviors and the risk for metabolic dysfunction-associated fatty liver disease (MAFLD). METHODS In this cross-sectional study of 5011 participants with self-reported sleep behaviors and radiologically diagnosed MAFLD, a comprehensive healthy sleep score was generated to evaluate the associations between sleep behaviors and MAFLD risk using multivariate logistic regression adjusting for demographics, lifestyles, medication, and metabolic comorbidities. Furthermore, mediation analysis was utilized to assess the extent to which obesity explains the effect of sleep quality on MAFLD risk. RESULTS Late bedtime, snoring, and daytime napping for over 30 minutes significantly associated with an increased risk of MAFLD, with odds ratios (OR) of 1.37 (95% CI 1.10, 1.70), 1.59 (95% CI 1.33, 1.91), and 1.17 (95% CI 1.02, 1.35), respectively, after full adjustments including obesity. Participants with disturbance in nighttime sleep and prolonged daytime napping showed the highest risk for MAFLD (OR 2.38, 95% CI 1.73, 3.27). Each additional increase of healthy sleep score was associated with a 16% reduction in MAFLD risk. Further stratified analysis revealed that people with a sedentary lifestyle and central obesity experienced more prominent adverse effects from poor sleep quality than others. Moreover, obesity accounted for only 20.77% of the total effect of sleep quality on MAFLD risk. CONCLUSIONS Sleep behaviors, both cumulatively and individually, are associated with MAFLD risk. Public health awareness and strategies should be encouraged to curb MAFLD.
Collapse
Affiliation(s)
- Jialu Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Shiyun Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Rui Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jingmeng Ju
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zhuoyu Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jichuan Shen
- Department of Toxicology, Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Minying Sun
- Department of Toxicology, Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Jiahua Fan
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Wei Zhu
- Department of Toxicology, Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Yan Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
19
|
Godoy-Lugo JA, Mendez DA, Rodriguez R, Nishiyama A, Nakano D, Soñanez-Organis JG, Ortiz RM. Improved lipogenesis gene expression in liver is associated with elevated plasma angiotensin 1-7 after AT1 receptor blockade in insulin-resistant OLETF rats. Mol Cell Endocrinol 2022; 555:111729. [PMID: 35921918 DOI: 10.1016/j.mce.2022.111729] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/19/2022]
Abstract
Increased angiotensin II (Ang II) signaling contributes to insulin resistance and liver steatosis. In addition to ameliorating hypertension, angiotensin receptor blockers (ARBs) improve lipid metabolism and hepatic steatosis, which are impaired with metabolic syndrome (MetS). Chronic blockade of the Ang II receptor type 1 (AT1) increases plasma angiotensin 1-7 (Ang 1-7), which mediates mechanisms counterregulatory to AT1 signaling. Elevated plasma Ang 1-7 is associated with decreased plasma triacylglycerol (TAG), cholesterol, glucose, and insulin; however, the benefits of RAS modulation to prevent non-alcoholic fatty liver disease (NAFLD) are not fully investigated. To better address the relationships among chronic ARB treatment, plasma Ang 1-7, and hepatic steatosis, three groups of 10-week-old-rats were studied: (1) untreated lean Long Evans Tokushima Otsuka (LETO), (2) untreated Otsuka Long Evans Tokushima Fatty (OLETF), and (3) OLETF + ARB (ARB; 10 mg olmesartan/kg/d × 6 weeks). Following overnight fasting, rats underwent an acute glucose load to better understand the dynamic metabolic responses during hepatic steatosis and early MetS. Tissues were collected at baseline (pre-load; T0) and 1 and 2 h post-glucose load. AT1 blockade increased plasma Ang 1-7 and decreased liver lipids, which was associated with decreased fatty acid transporter 5 (FATP5) and fatty acid synthase (FASN) expression. AT1 blockade decreased liver glucose and increased glucokinase (GCK) expression. These results demonstrate that during MetS, overactivation of AT1 promotes hepatic lipid deposition that is stimulated by an acute glucose load and lipogenesis genes, suggesting that the chronic hyperglycemia associated with MetS contributes to fatty liver pathologies via an AT1-mediated mechanism.
Collapse
Affiliation(s)
- Jose A Godoy-Lugo
- School of Natural Sciences, University of California, Merced, CA, USA.
| | - Dora A Mendez
- School of Natural Sciences, University of California, Merced, CA, USA
| | - Ruben Rodriguez
- School of Natural Sciences, University of California, Merced, CA, USA
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan
| | - Daisuke Nakano
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan
| | - Jose G Soñanez-Organis
- Universidad de Sonora, Departamento de Ciencias Químico Biológicas y Agropecuarias, Navojoa, Sonora, Mexico
| | - Rudy M Ortiz
- School of Natural Sciences, University of California, Merced, CA, USA
| |
Collapse
|
20
|
Guo J, Yu X, Liu Y, Lu L, Zhu D, Zhang Y, Li L, Zhang P, Gao Q, Lu X, Sun M. Prenatal hypothyroidism diminished exogenous NO-mediated diastolic effects in fetal rat thoracic aorta smooth muscle via increased oxidative stress. Reprod Toxicol 2022; 113:52-61. [PMID: 35970333 DOI: 10.1016/j.reprotox.2022.08.009] [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: 05/12/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Maternal hypothyroidism is an important problem of modern healthcare and is reported to increase the risk of cardiovascular diseases in the offspring later in life. However, it is unclear whether hypothyroidism during pregnancy causes vascular damage in the fetal period. We established the prenatal hypothyroidism rat model and collected the fetuses at the 21th day of gestation (GD21). Thyroid hormone concentrations in maternal and offspring blood serum were assessed by enzyme-linked immunosorbent assay (ELISA). The thoracic aortas of the fetuses were isolated for microvessel functional testing and histochemical stainings. qPCR and Western blot were performed to access mRNA and protein expression. We found that the concentrations of thyroid hormones in the serum of pregnant rats and fetuses were significantly suppressed at GD21. The responses of the fetal thoracic aortas to SNP were significantly attenuated in the PTU group. However, no statistical difference was found between the two groups when treated with either inhibitor (ODQ) or activator (BAY58-2667) of sGC. The production of O2-• in the arterial wall was significantly increased in hypothyroid fetuses. Moreover, the level of NADPH oxidase (NOX) was increased, while superoxide dismutase 2 (SOD2) was down-regulated in the PTU group, ultimately contributing to the increased production of superoxide. Additionally, decreased SNP-mediated vasodilation found in fetal vessels was improved by either NOX inhibitor (Apocynin) or SOD mimic (Tempol). These results indicate that increased oxidative stress is probably the cause of the diminished diastolic effect of exogenous NO in the thoracic artery of prenatal hypothyroidism exposed fetuses.
Collapse
Affiliation(s)
- Jun Guo
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Xi Yu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Yanping Liu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Likui Lu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Dan Zhu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Yingying Zhang
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Lingjun Li
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Pengjie Zhang
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Qinqin Gao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China
| | - Xiyuan Lu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China.
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu 215006, China.
| |
Collapse
|
21
|
Rotariu D, Babes EE, Tit DM, Moisi M, Bustea C, Stoicescu M, Radu AF, Vesa CM, Behl T, Bungau AF, Bungau SG. Oxidative stress - Complex pathological issues concerning the hallmark of cardiovascular and metabolic disorders. Biomed Pharmacother 2022; 152:113238. [PMID: 35687909 DOI: 10.1016/j.biopha.2022.113238] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 12/07/2022] Open
Abstract
Oxidative stress is a complex biological process characterized by the excessive production of reactive oxygen species (ROS) that act as destroyers of the REDOX balance in the body and, implicitly, inducing oxidative damage. All the metabolisms are impaired in oxidative stress and even nucleic acid balance is influenced. ROS will promote structural changes of the tissues and organs due to interaction with proteins and phospholipids. The constellation of the cardiovascular risk factors (CVRFs) will usually develop in subjects with predisposition to cardiac disorders. Oxidative stress is usually related with hypertension (HTN), diabetes mellitus (DM), obesity and cardiovascular diseases (CVDs) like coronary artery disease (CAD), cardiomyopathy or heart failure (HF), that can develop in subjects with the above-mentioned diseases. Elements describing the complex relationship between CVD and oxidative stress should be properly explored and described because prevention may be the optimal approach. Our paper aims to expose in detail the complex physiopathology of oxidative stress in CVD occurrence and novelties regarding the phenomenon. Biomarkers assessing oxidative stress or therapy targeting specific pathways represent a major progress that actually change the outcome of subjects with CVD. New antioxidants therapy specific for each CVD represents a captivating and interesting future perspective with tremendous benefits on subject's outcome.
Collapse
Affiliation(s)
- Dragos Rotariu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania.
| | - Emilia Elena Babes
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Delia Mirela Tit
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| | - Madalina Moisi
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Cristiana Bustea
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Manuela Stoicescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Andrei-Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India.
| | | | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| |
Collapse
|
22
|
Attia H, Albekairi N, Albdeirat L, Soliman A, Rajab R, Alotaibi H, Ali R, Badr A. Chrysin Attenuates Fructose-Induced Nonalcoholic Fatty Liver in Rats via Antioxidant and Anti-Inflammatory Effects: The Role of Angiotensin-Converting Enzyme 2/Angiotensin (1-7)/Mas Receptor Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9479456. [PMID: 35720181 PMCID: PMC9200559 DOI: 10.1155/2022/9479456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/12/2022] [Indexed: 11/25/2022]
Abstract
AIM Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of metabolic syndrome, and if untreated, it may propagate into end-stage liver disease. The classical arm of the renin-angiotensin system (RAS) has a fundamental role in triggering oxidative stress and inflammation, which play potential roles in the pathogenesis of NAFLD. However, the nonclassical alternative axis of RAS, angiotensin- (Ang-) converting enzyme 2 (ACE2)/Ang (1-7)/Mas receptor, opposes the actions of the classical arm, mitigates the metabolic dysfunction, and improves hepatic lipid metabolism rendering it a promising protective target against NAFLD. The current study is aimed at investigating the impact of chrysin, a well-known antioxidant flavonoid, on this defensive RAS axis in NAFLD. METHODS Rats were randomly distributed and treated daily for eight weeks as follows: the normal control, chrysin control (50 mg/kg, p.o), NAFLD group (received 20% fructose in drinking water), and treated groups (25 and 50 mg/kg chrysin given orally and concomitantly with fructose). Diminazene aceturate (DIZE) (15 mg/kg, s.c.) was used as a reference ACE2 activator. Key Findings. High fructose induced significant weight gain, hepatocyte degeneration with fat accumulation, and inflammatory cell infiltration (as examined by H&E staining). This was accompanied by a substantial increase in liver enzymes, glucose, circulating and hepatic triglycerides, lipid peroxides, inflammatory cytokines, and Ang II (the main component of classical RAS). At the same time, protein levels of ACE2, Ang (1-7), and Mas receptors were markedly reduced. Chrysin (25 and 50 mg/kg) significantly ameliorated these abnormalities, with a prominent effect of the dose of 50 mg/kg over DIZE and the lower dose in improving ACE2, Ang (1-7), and Mas. Significance. Chrysin is a promising efficient protective remedy against NAFLD; mechanisms include the activation of ACE2/Ang (1-7)/Mas axis.
Collapse
Affiliation(s)
- Hala Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
- Department of Biochemistry, College of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Norah Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Layal Albdeirat
- College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Arwa Soliman
- College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Reem Rajab
- College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Hend Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Rehab Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Amira Badr
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
- Department of Pharmacology and Toxicology, College of Pharmacy, Ain Shams University, Heliopolis, Cairo, Egypt
| |
Collapse
|
23
|
Ru L, Wang XM, Niu JQ. The miR-23-27-24 cluster: an emerging target in NAFLD pathogenesis. Acta Pharmacol Sin 2022; 43:1167-1179. [PMID: 34893685 PMCID: PMC9061717 DOI: 10.1038/s41401-021-00819-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022]
Abstract
The incidence of non-alcoholic fatty liver disease (NAFLD) is increasing globally, being the most widespread form of chronic liver disease in the west. NAFLD includes a variety of disease states, the mildest being non-alcoholic fatty liver that gradually progresses to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and eventually hepatocellular carcinoma. Small non-coding single-stranded microRNAs (miRNAs) regulate gene expression at the miRNA or translational level. Numerous miRNAs have been shown to promote NAFLD pathogenesis and progression through increasing lipid accumulation, oxidative stress, mitochondrial damage, and inflammation. The miR-23-27-24 clusters, composed of miR-23a-27a-24-2 and miR-23b-27b-24-1, have been implicated in various biological processes as well as many diseases. Herein, we review the current knowledge on miR-27, miR-24, and miR-23 in NAFLD pathogenesis and discuss their potential significance in NAFLD diagnosis and therapy.
Collapse
Affiliation(s)
- Lin Ru
- grid.430605.40000 0004 1758 4110Department of Hepatology, The First Hospital of Jilin University, Changchun, 130021 China
| | - Xiao-mei Wang
- grid.430605.40000 0004 1758 4110Department of Hepatology, The First Hospital of Jilin University, Changchun, 130021 China ,grid.430605.40000 0004 1758 4110Key Laboratory of Zoonosis Research, Ministry of Education, The First Hospital of Jilin University, Changchun, 130021 China
| | - Jun-qi Niu
- grid.430605.40000 0004 1758 4110Department of Hepatology, The First Hospital of Jilin University, Changchun, 130021 China ,grid.430605.40000 0004 1758 4110Key Laboratory of Zoonosis Research, Ministry of Education, The First Hospital of Jilin University, Changchun, 130021 China
| |
Collapse
|
24
|
Mahmudpour M, Vahdat K, Keshavarz M, Nabipour I. The COVID-19-diabetes mellitus molecular tetrahedron. Mol Biol Rep 2022; 49:4013-4024. [PMID: 35067816 PMCID: PMC8784222 DOI: 10.1007/s11033-021-07109-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/17/2021] [Indexed: 01/08/2023]
Abstract
Accumulating molecular evidence suggests that insulin resistance, rather than SARS-CoV-2- provoked beta-cell impairment, plays a major role in the observed rapid metabolic deterioration in diabetes, or new-onset hyperglycemia, during the COVID-19 clinical course. In order to clarify the underlying complexity of COVID-19 and diabetes mellitus interactions, we propose the imaginary diabetes-COVID-19 molecular tetrahedron with four lateral faces consisting of SARS-CoV-2 entry via ACE2 (lateral face 1), the viral hijacking and replication (lateral face 2), acute inflammatory responses (lateral face 3), and the resulting insulin resistance (lateral face 4). The entrance of SARS-CoV-2 using ACE2 receptor triggers an array of multiple molecular signaling beyond that of the angiotensin II/ACE2-Ang-(1-7) axis, such as down-regulation of PGC-1 α/irisin, increased SREBP-1c activity, upregulation of CD36 and Sirt1 inhibition leading to insulin resistance. In another arm of the molecular cascade, the SARS-CoV-2 hijacking and replication induces a series of molecular events in the host cell metabolic machinery, including upregulation of SREBP-2, decrement in Sirt1 expression, dysregulation in PPAR-ɣ, and LPI resulting in insulin resistance. The COVID-19-diabetes molecular tetrahedron may suggest novel targets for therapeutic interventions to overcome insulin resistance that underlies the pathophysiology of worsening metabolic control in patients with diabetes mellitus or the new-onset of hyperglycemia in COVID-19.
Collapse
Affiliation(s)
- Mehdi Mahmudpour
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Katayoun Vahdat
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Iraj Nabipour
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran.
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran.
| |
Collapse
|
25
|
Godoy-Lugo JA, Thorwald MA, Hui DY, Nishiyama A, Nakano D, Soñanez-Organis JG, Ortiz RM. Chronic angiotensin receptor activation promotes hepatic triacylglycerol accumulation during an acute glucose challenge in obese-insulin-resistant OLETF rats. Endocrine 2022; 75:92-107. [PMID: 34327606 PMCID: PMC8763929 DOI: 10.1007/s12020-021-02834-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/18/2021] [Indexed: 11/01/2022]
Abstract
PURPOSE Angiotensin receptor blockers (ARBs) can ameliorate metabolic syndrome (MetS)-associated dyslipidemia, hepatic steatosis, and glucose intolerance, suggesting that angiotensin receptor (AT1) over-activation contributes to impaired lipid and glucose metabolism, which is characteristic of MetS. The aim of this study was to evaluate changes in the lipid profile and proteins of fatty acid uptake, triacylglycerol (TAG) synthesis, and β-oxidation to better understand the links between AT1 overactivation and non-alcoholic fatty liver disease (NAFLD) during MetS. METHODS Four groups of 25-week-old-rats were used: (1) untreated LETO, (2) untreated OLETF, (3) OLETF + angiotensin receptor blocker (ARB; 10 mg olmesartan/kg/d × 8 weeks) and (4) OLETF ± ARB (MINUS; 10 mg olmesartan/kg/d × 4 weeks, then removed until dissection). To investigate the dynamic shifts in metabolism, animals were dissected after an oral glucose challenge (fasting, 3 and 6 h post-glucose). RESULTS Compared to OLETF, plasma total cholesterol and TAG remained unchanged in ARB. However, liver TAG was 55% lesser in ARB than OLETF, and remained lower throughout the challenge. Basal CD36 and ApoB were 28% and 29% lesser, respectively, in ARB than OLETF. PRDX6 abundance in ARB was 45% lesser than OLETF, and it negatively correlated with liver TAG in ARB. CONCLUSIONS Chronic blockade of AT1 protects the liver from TAG accumulation during glucose overload. This may be achieved by modulating NEFA uptake and increasing TAG export via ApoB. Our study highlights the contributions of AT1 signaling to impaired hepatic substrate metabolism and the detriments of a high-glucose load and its potential contribution to steatosis during MetS.
Collapse
Affiliation(s)
- Jose A Godoy-Lugo
- School of Natural Sciences, University of California, Merced, CA, USA.
| | - Max A Thorwald
- School of Natural Sciences, University of California, Merced, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - David Y Hui
- Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan
| | - Daisuke Nakano
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan
| | - Jose G Soñanez-Organis
- Universidad de Sonora, Departamento de Ciencias Químico Biológicas y Agropecuarias, Navojoa, Sonora, Mexico
| | - Rudy M Ortiz
- School of Natural Sciences, University of California, Merced, CA, USA
| |
Collapse
|
26
|
Sabri S, Bourron O, Phan F, Nguyen LS. Interactions between diabetes and COVID-19: A narrative review. World J Diabetes 2021; 12:1674-1692. [PMID: 34754370 PMCID: PMC8554367 DOI: 10.4239/wjd.v12.i10.1674] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/29/2021] [Accepted: 08/31/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes, whether due to pancreatic beta cells insufficiency or peripheral resistance to insulin, has been suggested as a risk factor of developing severe acute respiratory disease coronavirus-2 (SARS-CoV-2) infections. Indeed, diabetes has been associated with a higher risk of infections and higher risk of developing severe forms of coronavirus disease 2019 (COVID-19) related pneumonia. Diabetic patients often present associated comorbidities such as obesity, hypertension and cardiovascular diseases, and complications of diabetes, including chronic kidney disease, vasculopathy and relative immune dysfunction, all of which make them more susceptible to infectious complications. Moreover, they often present low-grade inflammation with increased circulating interleukin levels, endothelial susceptibility to inflammation and dysfunction, and finally, hyperglycemia, which increases this risk. Additionally, corticosteroids, which count among the few medications which showed benefit on survival and mechanical ventilation requirement in COVID-19 pneumonia in large randomized controlled trials, are associated to new onsets of diabetes, and metabolic disorders in patients with previous history of diabetes. Finally, SARS-CoV-2 via the alternate effects of the renin-angiotensin system, mediated by the angiotensin-converting-enzyme 2, was also associated with insulin resistance in key tissues involved in glucose homeostasis, such as liver, skeletal muscles, and adipose tissue; and also, with impaired insulin secretion by pancreatic β-cells. In this work, we reviewed all elements which may help understand how diabetes affects patients with COVID-19, how treatments affect outcomes in patients with COVID-19, how they may cause new onsets of diabetes, and finally review how SARS-CoV-2 may inherently be a risk factor of developing diabetes, through immune-mediated diabetogenic mechanisms.
Collapse
Affiliation(s)
- Sophia Sabri
- Intensive Care Medicine, CMC Ambroise Paré, Neuilly-Sur-Seine 92200, France
| | - Olivier Bourron
- Sorbonne Université Médecine; Assistance publique Hôpitaux de Paris (APHP), Service de Diabétologie, Hôpital Pitié-Salpêtrière; INSERM UMRS_1138, Centre de recherche des Cordeliers; Institute of CArdiometabolisme and Nutrition (ICAN), Paris 75013, France
| | - Franck Phan
- Sorbonne Université Médecine; Assistance publique Hôpitaux de Paris (APHP), Service de Diabétologie, Hôpital Pitié-Salpêtrière; INSERM UMRS_1138, Centre de recherche des Cordeliers; Institute of CArdiometabolisme and Nutrition (ICAN), Paris 75013, France
| | - Lee S Nguyen
- Research and Innovation, RICAP, CMC Ambroise Paré, Neuilly-Sur-Seine 92200, France
| |
Collapse
|
27
|
Stearoyl-CoA Desaturase (SCD) Induces Cardiac Dysfunction with Cardiac Lipid Overload and Angiotensin II AT1 Receptor Protein Up-Regulation. Int J Mol Sci 2021; 22:ijms22189883. [PMID: 34576047 PMCID: PMC8472087 DOI: 10.3390/ijms22189883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/17/2022] Open
Abstract
Heart failure is a major cause of death worldwide with insufficient treatment options. In the search for pathomechanisms, we found up-regulation of an enzyme, stearoyl-CoA desaturase 1 (Scd1), in different experimental models of heart failure induced by advanced atherosclerosis, chronic pressure overload, and/or volume overload. Because the pathophysiological role of Scd1/SCD in heart failure is not clear, we investigated the impact of cardiac SCD upregulation through the generation of C57BL/6-Tg(MHCSCD)Sjaa mice with myocardium-specific expression of SCD. Echocardiographic examination showed that 4.9-fold-increased SCD levels triggered cardiac hypertrophy and symptoms of heart failure at an age of eight months. Tg-SCD mice had a significantly reduced left ventricular cardiac ejection fraction of 25.7 ± 2.9% compared to 54.3 ± 4.5% of non-transgenic B6 control mice. Whole-genome gene expression profiling identified up-regulated heart-failure-related genes such as resistin, adiponectin, and fatty acid synthase, and type 1 and 3 collagens. Tg-SCD mice were characterized by cardiac lipid accumulation with 1.6- and 1.7-fold-increased cardiac contents of saturated lipids, palmitate, and stearate, respectively. In contrast, unsaturated lipids were not changed. Together with saturated lipids, apoptosis-enhancing p53 protein contents were elevated. Imaging by autoradiography revealed that the heart-failure-promoting and membrane-spanning angiotensin II AT1 receptor protein of Tg-SCD hearts was significantly up-regulated. In transfected HEK cells, the expression of SCD increased the number of cell-surface angiotensin II AT1 receptor binding sites. In addition, increased AT1 receptor protein levels were detected by fluorescence spectroscopy of fluorescent protein-labeled AT1 receptor-Cerulean. Taken together, we found that SCD promotes cardiac dysfunction with overload of cardiotoxic saturated lipids and up-regulation of the heart-failure-promoting AT1 receptor protein.
Collapse
|
28
|
Abstract
Introduction: Oxidative stress underlies the pathophysiology of various etiologies of chronic liver disease and contributes to the development of hepatocarcinogenesis.Areas covered: This review focuses on the impact of oxidative stress in various etiologies of chronic liver disease such as alcoholic liver disease (ALD), nonalcoholic steatohepatitis (NASH), hepatitis B virus (HBV), and hepatitis C virus (HCV) infection. The efficacy of antioxidants in laboratory, animal, and clinical studies in chronic liver disease is also reviewed.Expert opinion: Currently, there are limited targeted pharmacotherapeutics for NASH and no pharmacotherapeutics for ALD and antioxidant supplementation may be useful in these conditions to improve liver function and reverse fibrosis. Antioxidants may also be used in patients with HBV or HCV infection to supplement antiviral therapies. Specific genotypes of antioxidant and prooxidant genes render patients more susceptible to liver cirrhosis and hepatocellular carcinoma while other individual characteristics like age, genotype, and metabolomic profiling can influence the efficacy of antioxidants on CLD. More research needs to be done to establish the safety, efficacy, and dosage of antioxidants and to establish the ideal patient profile that will benefit the most from antioxidant treatment.
Collapse
Affiliation(s)
- Sophia Seen
- Tan Tock Seng Hospital, Singapore, Singapore
| |
Collapse
|
29
|
Laghlam D, Jozwiak M, Nguyen LS. Renin-Angiotensin-Aldosterone System and Immunomodulation: A State-of-the-Art Review. Cells 2021; 10:cells10071767. [PMID: 34359936 PMCID: PMC8303450 DOI: 10.3390/cells10071767] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
The renin–angiotensin system (RAS) has long been described in the field of cardiovascular physiology as the main player in blood pressure homeostasis. However, other effects have since been described, and include proliferation, fibrosis, and inflammation. To illustrate the immunomodulatory properties of the RAS, we chose three distinct fields in which RAS may play a critical role and be the subject of specific treatments. In oncology, RAS hyperactivation has been associated with tumor migration, survival, cell proliferation, and angiogenesis; preliminary data showed promise of the benefit of RAS blockers in patients treated for certain types of cancer. In intensive care medicine, vasoplegic shock has been associated with severe macro- and microcirculatory imbalance. A relative insufficiency in angiotensin II (AngII) was associated to lethal outcomes and synthetic AngII has been suggested as a specific treatment in these cases. Finally, in solid organ transplantation, both AngI and AngII have been associated with increased rejection events, with a regional specificity in the RAS activity. These elements emphasize the complexity of the direct and indirect interactions of RAS with immunomodulatory pathways and warrant further research in the field.
Collapse
|
30
|
Stachowicz A, Wiśniewska A, Kuś K, Białas M, Łomnicka M, Totoń-Żurańska J, Kiepura A, Stachyra K, Suski M, Bujak-Giżycka B, Jawień J, Olszanecki R. Diminazene Aceturate Stabilizes Atherosclerotic Plaque and Attenuates Hepatic Steatosis in apoE-Knockout Mice by Influencing Macrophages Polarization and Taurine Biosynthesis. Int J Mol Sci 2021; 22:5861. [PMID: 34070749 PMCID: PMC8199145 DOI: 10.3390/ijms22115861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis and nonalcoholic fatty liver disease are leading causes of morbidity and mortality in the Western countries. The renin-angiotensin system (RAS) with its two main opposing effectors, i.e., angiotensin II (Ang II) and Ang-(1-7), is widely recognized as a major regulator of cardiovascular function and body metabolic processes. Angiotensin-converting enzyme 2 (ACE2) by breaking-down Ang II forms Ang-(1-7) and thus favors Ang-(1-7) actions. Therefore, the aim of our study was to comprehensively evaluate the influence of prolonged treatment with ACE2 activator, diminazene aceturate (DIZE) on the development of atherosclerotic lesions and hepatic steatosis in apoE-/- mice fed a high-fat diet (HFD). We have shown that DIZE stabilized atherosclerotic lesions and attenuated hepatic steatosis in apoE-/- mice fed an HFD. Such effects were associated with decreased total macrophages content and increased α-smooth muscle actin levels in atherosclerotic plaques. Moreover, DIZE changed polarization of macrophages towards increased amount of anti-inflammatory M2 macrophages in the atherosclerotic lesions. Interestingly, the anti-steatotic action of DIZE in the liver was related to the elevated levels of HDL in the plasma, decreased levels of triglycerides, and increased biosynthesis and concentration of taurine in the liver of apoE-/- mice. However, exact molecular mechanisms of both anti-atherosclerotic and anti-steatotic actions of DIZE require further investigations.
Collapse
Affiliation(s)
- Aneta Stachowicz
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Anna Wiśniewska
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Katarzyna Kuś
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Magdalena Białas
- Chair of Pathomorphology, Jagiellonian University Medical College, 31-531 Krakow, Poland;
| | - Magdalena Łomnicka
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Justyna Totoń-Żurańska
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Anna Kiepura
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Kamila Stachyra
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Maciej Suski
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Beata Bujak-Giżycka
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Jacek Jawień
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Rafał Olszanecki
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| |
Collapse
|
31
|
Yang YJ, Kim DJ. An Overview of the Molecular Mechanisms Contributing to Musculoskeletal Disorders in Chronic Liver Disease: Osteoporosis, Sarcopenia, and Osteoporotic Sarcopenia. Int J Mol Sci 2021; 22:2604. [PMID: 33807573 PMCID: PMC7961345 DOI: 10.3390/ijms22052604] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
The prevalence of osteoporosis and sarcopenia is significantly higher in patients with liver disease than in those without liver disease and osteoporosis and sarcopenia negatively influence morbidity and mortality in liver disease, yet these musculoskeletal disorders are frequently overlooked in clinical practice for patients with chronic liver disease. The objective of this review is to provide a comprehensive understanding of the molecular mechanisms of musculoskeletal disorders accompanying the pathogenesis of liver disease. The increased bone resorption through the receptor activator of nuclear factor kappa (RANK)-RANK ligand (RANKL)-osteoprotegerin (OPG) system and upregulation of inflammatory cytokines and decreased bone formation through increased bilirubin and sclerostin and lower insulin-like growth factor-1 are important mechanisms for osteoporosis in patients with liver disease. Sarcopenia is associated with insulin resistance and obesity in non-alcoholic fatty liver disease, whereas hyperammonemia, low amount of branched chain amino acids, and hypogonadism contributes to sarcopenia in liver cirrhosis. The bidirectional crosstalk between muscle and bone through myostatin, irisin, β-aminoisobutyric acid (BAIBA), osteocalcin, as well as the activation of the RANK and the Wnt/β-catenin pathways are associated with osteosarcopenia. The increased understandings for these musculoskeletal disorders would be contributes to the development of effective therapies targeting the pathophysiological mechanism involved.
Collapse
Affiliation(s)
- Young Joo Yang
- Department of Internal Medicine, Hallym University College of Medicine, Gangwon-do, Chuncheon 24252, Korea;
- Institute for Liver and Digestive Diseases, Hallym University, Gangwon-do, Chuncheon 24253, Korea
| | - Dong Joon Kim
- Department of Internal Medicine, Hallym University College of Medicine, Gangwon-do, Chuncheon 24252, Korea;
- Institute for Liver and Digestive Diseases, Hallym University, Gangwon-do, Chuncheon 24253, Korea
| |
Collapse
|
32
|
ACE2 and energy metabolism: the connection between COVID-19 and chronic metabolic disorders. Clin Sci (Lond) 2021; 135:535-554. [PMID: 33533405 DOI: 10.1042/cs20200752] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022]
Abstract
The renin-angiotensin system (RAS) has currently attracted increasing attention due to its potential function in regulating energy homeostasis, other than the actions on cellular growth, blood pressure, fluid, and electrolyte balance. The existence of RAS is well established in metabolic organs, including pancreas, liver, skeletal muscle, and adipose tissue, where activation of angiotensin-converting enzyme (ACE) - angiotensin II pathway contributes to the impairment of insulin secretion, glucose transport, fat distribution, and adipokines production. However, the activation of angiotensin-converting enzyme 2 (ACE2) - angiotensin (1-7) pathway, a novel branch of the RAS, plays an opposite role in the ACE pathway, which could reverse these consequences by improving local microcirculation, inflammation, stress state, structure remolding, and insulin signaling pathway. In addition, new studies indicate the protective RAS arm possesses extraordinary ability to enhance brown adipose tissue (BAT) activity and induces browning of white adipose tissue, and consequently, it leads to increased energy expenditure in the form of heat instead of ATP synthesis. Interestingly, ACE2 is the receptor of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is threating public health worldwide. The main complications of SARS-CoV-2 infected death patients include many energy metabolism-related chronic diseases, such as diabetes. The specific mechanism leading to this phenomenon is largely unknown. Here, we summarize the latest pharmacological and genetic tools on regulating ACE/ACE2 balance and highlight the beneficial effects of the ACE2 pathway axis hyperactivity on glycolipid metabolism, as well as the thermogenic modulation.
Collapse
|
33
|
Kim KM, Roh JH, Lee S, Yoon JH. Clinical implications of renin-angiotensin system inhibitors for development and progression of non-alcoholic fatty liver disease. Sci Rep 2021; 11:2884. [PMID: 33536442 PMCID: PMC7858633 DOI: 10.1038/s41598-021-81959-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
Recently, there has been an increasing interest in the therapeutic efficacy of RAS inhibitors (RASi) in patients with non-alcoholic fatty liver disease (NAFLD) because they may reduce oxidative stress, inflammatory markers, and enhanced fibrosis. An objective of this study was to investigate the role of RASi on NAFLD development and progression in a large cohort. We conducted a nested case-control study. Study subjects were classified into two study cohorts according to baseline NAFLD status: non-NAFLD (n = 184,581) and established NALFD (n = 27,565). An NAFLD development or progression case was defined as a patient with newly developed NAFLD or new progression of advanced fibrosis from non-NAFLD and established NALFD cohorts, respectively. A conditional logistic regression analysis was conducted to estimate the associations between RASi exposure and NAFLD development/progression. Overall, no significant association was evident between RASi use and NAFLD development or progression (NAFLD development; ever-user vs. never-user: OR 1.017; 95% CI 0.842-1.230, NAFLD progression; ever-user vs. never-user: aOR 0.942; 95% CI 0.803-1.105). RASi ever-use in cases of individuals who were obese or who had normal fasting plasma glucose (FPG) was associated with reduced risk of both NAFLD development (body mass index (BMI) ≥ 25 kg/m2: 0.708 [95% confidence interval (CI) 0.535-0.937], FPG of < 100 mg/mL: 0.774 [95% CI 0.606-0.987]) and progression (BMI ≥ 25 kg/m2: 0.668 [95% CI 0.568-0.784], FPG of < 100 mg/mL: 0.732 [95% CI 0.582-0.921]). The present study did not verify a significant overall association between RASi use and NAFLD development/progression but suggested that RASi might prevent NAFLD development and progression among specific subjects.
Collapse
Affiliation(s)
- Kwang Min Kim
- Department of Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, South Korea
| | - Ji-Hye Roh
- College of Pharmacy, Pusan National University, Busan, South Korea
| | - Sangjin Lee
- Department of Statistics, College of Natural Science, Pusan National University, Busan, South Korea
| | - Jeong-Hyun Yoon
- College of Pharmacy, Pusan National University, Busan, South Korea.
| |
Collapse
|
34
|
Pola A, Murthy KS, Santhekadur PK. COVID-19 and gastrointestinal system: A brief review. Biomed J 2021; 44:245-251. [PMID: 34130944 PMCID: PMC7832457 DOI: 10.1016/j.bj.2021.01.001] [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: 08/10/2020] [Revised: 12/25/2020] [Accepted: 01/11/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is a recent pandemic that is still a major health problem of modern times and already more than 17.5 lakhs people succumbed to this deadly disease. This disease is caused by novel coronavirus which is named SARS-COV-2 by the International Committee on Taxonomy of Viruses. This virus originated from Wuhan city in Hubei province of China in December 2019 and within a short period spread across the many countries in the globe. There are a lot of basic as well as clinical research is going on to study the mode of transmission and the mechanism of action of SARS-COV-2 infection and its therapeutics. SARS-COV-2 is not only known to infect lungs, but it also infects other organs in the human body including the gastrointestinal (GI) tract, the liver, and the pancreas via the angiotensin-converting enzyme (ACE) 2, an important component of the renin-angiotensin system. In this short review, we are mainly discussing the mode of SARS-COV-2 transmission, physiological counterbalancing roles of ACE2 and ACE and the tissue patterns of ACE2 expression, and the overall effect of COVID19 on human gastrointestinal System. Therefore, this review sheds light on the possible mechanism of SARS-COV-2 infection in the GI system and its pathological symptoms raising a potential possibility of GI tract acting as a secondary site for SARS-CoV-2 tropism and infection. Finally, future studies to understand the fecal-oral transmission of the virus and the correlation of viral load and severity of GI symptoms are proposed to gain knowledge of the GI symptoms in COVID-19 to aid in early diagnosis and prognosis.
Collapse
Affiliation(s)
- Akhil Pola
- Department of Biochemistry, Center of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| | - Karnam S Murthy
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Prasanna K Santhekadur
- Department of Biochemistry, Center of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, Karnataka, India.
| |
Collapse
|
35
|
Fernández-Mincone T, Contreras-Briceño F, Espinosa-Ramírez M, García-Valdés P, López-Fuenzalida A, Riquelme A, Arab JP, Cabrera D, Arrese M, Barrera F. Nonalcoholic fatty liver disease and sarcopenia: pathophysiological connections and therapeutic implications. Expert Rev Gastroenterol Hepatol 2020; 14:1141-1157. [PMID: 32811209 DOI: 10.1080/17474124.2020.1810563] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Nonalcoholic fatty liver disease (NAFLD) is currently one of the most common liver diseases worldwide. Recent data suggest that loss of skeletal muscle mass and function (i.e. sarcopenia) is highly prevalent and frequently overlooked in NAFLD patients. Experimental and clinical data suggest that the relationship between NAFLD and sarcopenia is pathophysiologically complex and bi-directional and there is a growing interest in unveiling how sarcopenia could influence NAFLD development and progression. AREAS COVERED PubMed/MEDLINE was searched for articles related to concomitant occurrence of NAFLD and sarcopenia between January 2013 and April 2020. Areas covered in this review include: (1) updated sarcopenia diagnosis strategy, (2) discussion of current data on pathophysiological connections between NAFLD and sarcopenia, and (3) analysis of current and future therapeutic implications of this knowledge. EXPERT OPINION Clinical studies describe a consistent association between NAFLD and sarcopenia, although a cause-effect relation remains to be determined. Active implementation of current diagnosis algorithms and optimized treatment can prevent sarcopenia related complications in subjects with NAFLD. Pathogenic pathways implicated in this relation are multiple and complex, a better understanding of them can provide novel biomarkers and targeted therapies that will hopefully have an important impact in NAFLD management.
Collapse
Affiliation(s)
- Tiziana Fernández-Mincone
- Laboratorio de Fisiología del Ejercicio, Departamento Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Felipe Contreras-Briceño
- Laboratorio de Fisiología del Ejercicio, Departamento Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Maximiliano Espinosa-Ramírez
- Laboratorio de Fisiología del Ejercicio, Departamento Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Patricio García-Valdés
- Laboratorio de Fisiología del Ejercicio, Departamento Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Antonio López-Fuenzalida
- Laboratorio de Fisiología del Ejercicio, Departamento Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Arnoldo Riquelme
- Departamento Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile.,Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Daniel Cabrera
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile.,Facultad de Ciencias Médicas, Universidad Bernardo O Higgins , Santiago, Chile
| | - Marco Arrese
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Francisco Barrera
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile
| |
Collapse
|
36
|
Song L, Liu J, Shi T, Zhang Y, Xin Z, Cao X, Yang J. Angiotensin‐(1‐7), the product of ACE2 ameliorates NAFLD by acting through its receptor Mas to regulate hepatic mitochondrial function and glycolipid metabolism. FASEB J 2020; 34:16291-16306. [PMID: 33078906 DOI: 10.1096/fj.202001639r] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/27/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Li‐Ni Song
- Beijing Key Laboratory of Diabetes Research and Care Department of Endocrinology Beijing Diabetes Institute Beijing Tongren Hospital Capital Medical University Beijing China
| | - Jing‐Yi Liu
- Beijing Key Laboratory of Diabetes Research and Care Department of Endocrinology Beijing Diabetes Institute Beijing Tongren Hospital Capital Medical University Beijing China
| | - Ting‐Ting Shi
- Beijing Key Laboratory of Diabetes Research and Care Department of Endocrinology Beijing Diabetes Institute Beijing Tongren Hospital Capital Medical University Beijing China
| | - Yi‐Chen Zhang
- Beijing Key Laboratory of Diabetes Research and Care Department of Endocrinology Beijing Diabetes Institute Beijing Tongren Hospital Capital Medical University Beijing China
| | - Zhong Xin
- Beijing Key Laboratory of Diabetes Research and Care Department of Endocrinology Beijing Diabetes Institute Beijing Tongren Hospital Capital Medical University Beijing China
| | - Xi Cao
- Beijing Key Laboratory of Diabetes Research and Care Department of Endocrinology Beijing Diabetes Institute Beijing Tongren Hospital Capital Medical University Beijing China
| | - Jin‐Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care Department of Endocrinology Beijing Diabetes Institute Beijing Tongren Hospital Capital Medical University Beijing China
| |
Collapse
|
37
|
Acquah S. Implications of COVID-19 Pandemic on Evolution of Diabetes in Malaria-Endemic African Region. J Diabetes Res 2020; 2020:8205261. [PMID: 33134395 PMCID: PMC7568783 DOI: 10.1155/2020/8205261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/08/2020] [Accepted: 09/26/2020] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic continues to cause havoc to many countries of the globe, with no end in sight, due to nonavailability of a given vaccine or treatment regimen. The pandemic has so far had a relatively limited impact on the African continent, which contributes more than 93% of global malaria burden. However, the limited burden of COVID-19 pandemic on the African region could have long-term implications on the health and wellbeing of affected inhabitants due to its malaria-endemic status. Malaria causes recurrent insulin resistance with episodes of infection at relatively low parasitaemia. Angiotensin-converting enzyme 2 (ACE2) which is widely distributed in the human body is implicated in the pathogenesis of malaria, type 2 diabetes mellitus (T2DM), and COVID-19. Use of ACE2 by the COVID-19 virus induces inflammation and oxidative stress, which can lead to insulin resistance. Although COVID-19 patients in malaria-endemic African region may not exhibit severe signs and symptoms of the disease, their risk of exhibiting heightened insulin resistance and possible future development of T2DM is high due to their prior exposure to malaria. African governments must double efforts at containing the continued spread of the virus without neglecting existing malarial control measures if the region is to avert the plausible long-term impact of the pandemic in terms of future development of T2DM.
Collapse
Affiliation(s)
- Samuel Acquah
- Department of Medical Biochemistry, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
38
|
Alshahrani S. Aliskiren – A promising antioxidant agent beyond hypertension reduction. Chem Biol Interact 2020; 326:109145. [DOI: 10.1016/j.cbi.2020.109145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/11/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
|
39
|
Cremonese C, Schierwagen R, Uschner FE, Torres S, Tyc O, Ortiz C, Schulz M, Queck A, Kristiansen G, Bader M, Sauerbruch T, Weiskirchen R, Walther T, Trebicka J, Klein S. Short-Term Western Diet Aggravates Non-Alcoholic Fatty Liver Disease (NAFLD) With Portal Hypertension in TGR(mREN2)27 Rats. Int J Mol Sci 2020; 21:E3308. [PMID: 32392802 PMCID: PMC7246932 DOI: 10.3390/ijms21093308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is gaining in importance and is linked to obesity. Especially, the development of fibrosis and portal hypertension in NAFLD patients requires treatment. Transgenic TGR(mREN2)27 rats overexpressing mouse renin spontaneously develop NAFLD with portal hypertension but without obesity. This study investigated the additional role of obesity in this model on the development of portal hypertension and fibrosis. Obesity was induced in twelve-week old TGR(mREN2)27 rats after receiving Western diet (WD) for two or four weeks. Liver fibrosis was assessed using standard techniques. Hepatic expression of transforming growth factor-β1 (TGF-β1), collagen type Iα1, α-smooth muscle actin, and the macrophage markers Emr1, as well as the chemoattractant Ccl2, interleukin-1β (IL1β) and tumor necrosis factor-α (TNFα) were analyzed. Assessment of portal and systemic hemodynamics was performed using the colored microsphere technique. As expected, WD induced obesity and liver fibrosis as confirmed by Sirius Red and Oil Red O staining. The expression of the monocyte-macrophage markers, Emr1, Ccl2, IL1β and TNFα were increased during feeding of WD, indicating infiltration of macrophages into the liver, even though this increase was statistically not significant for the EGF module-containing mucin-like receptor (Emr1) mRNA expression levels. Of note, portal pressure increased with the duration of WD compared to animals that received a normal chow. Besides obesity, WD feeding increased systemic vascular resistance reflecting systemic endothelial and splanchnic vascular dysfunction. We conclude that transgenic TGR(mREN2)27 rats are a suitable model to investigate NAFLD development with liver fibrosis and portal hypertension. Tendency towards elevated expression of Emr1 is associated with macrophage activity point to a significant role of macrophages in NAFLD pathogenesis, probably due to a shift of the renin-angiotensin system towards a higher activation of the classical pathway. The hepatic injury induced by WD in TGR(mREN2)27 rats is suitable to evaluate different stages of fibrosis and portal hypertension in NAFLD with obesity.
Collapse
Affiliation(s)
- Carla Cremonese
- Department of Internal Medicine I, Goethe University Frankfurt, 60323 Frankfurt, Germany; (C.C.); (R.S.); (F.E.U.); (S.T.); (O.T.); (C.O.); (M.S.); (A.Q.); (S.K.)
| | - Robert Schierwagen
- Department of Internal Medicine I, Goethe University Frankfurt, 60323 Frankfurt, Germany; (C.C.); (R.S.); (F.E.U.); (S.T.); (O.T.); (C.O.); (M.S.); (A.Q.); (S.K.)
| | - Frank Erhard Uschner
- Department of Internal Medicine I, Goethe University Frankfurt, 60323 Frankfurt, Germany; (C.C.); (R.S.); (F.E.U.); (S.T.); (O.T.); (C.O.); (M.S.); (A.Q.); (S.K.)
| | - Sandra Torres
- Department of Internal Medicine I, Goethe University Frankfurt, 60323 Frankfurt, Germany; (C.C.); (R.S.); (F.E.U.); (S.T.); (O.T.); (C.O.); (M.S.); (A.Q.); (S.K.)
| | - Olaf Tyc
- Department of Internal Medicine I, Goethe University Frankfurt, 60323 Frankfurt, Germany; (C.C.); (R.S.); (F.E.U.); (S.T.); (O.T.); (C.O.); (M.S.); (A.Q.); (S.K.)
| | - Cristina Ortiz
- Department of Internal Medicine I, Goethe University Frankfurt, 60323 Frankfurt, Germany; (C.C.); (R.S.); (F.E.U.); (S.T.); (O.T.); (C.O.); (M.S.); (A.Q.); (S.K.)
| | - Martin Schulz
- Department of Internal Medicine I, Goethe University Frankfurt, 60323 Frankfurt, Germany; (C.C.); (R.S.); (F.E.U.); (S.T.); (O.T.); (C.O.); (M.S.); (A.Q.); (S.K.)
| | - Alexander Queck
- Department of Internal Medicine I, Goethe University Frankfurt, 60323 Frankfurt, Germany; (C.C.); (R.S.); (F.E.U.); (S.T.); (O.T.); (C.O.); (M.S.); (A.Q.); (S.K.)
| | - Glen Kristiansen
- Institute for Pathology, University of Bonn, 53127 Bonn, Germany;
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine, 13092 Berlin, Germany;
| | - Tilman Sauerbruch
- Department of Internal Medicine I, University Hospital of Bonn, 53127 Bonn, Germany;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany;
| | - Thomas Walther
- Department of Pharmacology and Therapeutics, University College Cork, T12 YN60 Cork, Ireland;
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, Goethe University Frankfurt, 60323 Frankfurt, Germany; (C.C.); (R.S.); (F.E.U.); (S.T.); (O.T.); (C.O.); (M.S.); (A.Q.); (S.K.)
- Institute for Bioengineering of Catalonia, 08028 Barcelona, Spain
- European Foundation for the Study of Chronic Liver Failure, 08021 Barcelona, Spain
- Faculty of Health Sciences, University of Southern Denmark, 5000 Odense, Denmark
| | - Sabine Klein
- Department of Internal Medicine I, Goethe University Frankfurt, 60323 Frankfurt, Germany; (C.C.); (R.S.); (F.E.U.); (S.T.); (O.T.); (C.O.); (M.S.); (A.Q.); (S.K.)
| |
Collapse
|
40
|
Rauchová H, Hojná S, Kadlecová M, Vaněčková I, Zicha J. Sex differences in blood pressure of aged Ren-2 transgenic rats. Physiol Res 2020; 69:245-252. [DOI: 10.33549/physiolres.934369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Sex-related differences were observed not only in human but also in experimental hypertension. The aim of our study was to compare blood pressure (BP) of aged male and female heterozygous transgenic rats (TGR) harboring Ren-2 mouse gene, with their normotensive Hannover Sprague-Dawley (HanSD) controls. At the age of 9 months, systolic (SBP) and diastolic blood pressure (DBP) were measured by a direct puncture of carotid artery in rats awaking from isoflurane anesthesia. Thiobarbituric acid-reactive species (TBARS) formation was monitored as indicator of lipid peroxidation damage in heart, kidney and liver, whereas intracellular content of reduced glutathione was determined in the same organs as the main intracellular antioxidant. Furthermore, plasma triglycerides and total cholesterol as well as high-density lipoprotein (HDL) and low-density lipoprotein (LDL) fractions of cholesterol were measured. As compared to HanSD rats, we found significantly elevated BP only in male TGR (MAP: 123±1 vs. 171±5, SBP: 150±2 vs. 208±7, and DBP: 99±3 vs. 140±4 mm Hg), but not between TGR and HanSD females, which were both normotensive. We also did not find any significant differences in TBARS and reduced glutathione in the three above mentioned organs as well as in plasma cholesterol or its HDL and LDL fractions between transgene-negative HanSD and TGR animals of either sex. However, we found significant sex differences in TBARS, glutathione and plasma lipids in both rat strains. Our results confirmed that aged TGR exhibit a marked sexual BP dimorphism, which does not seem to be dependent on oxidative stress or abnormal cholesterol metabolism.
Collapse
Affiliation(s)
- H. Rauchová
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | | | | | |
Collapse
|
41
|
Romão MH, de Bem GF, Santos IB, de Andrade Soares R, Ognibene DT, de Moura RS, da Costa CA, Resende ÂC. Açaí (Euterpe oleracea Mart.) seed extract protects against hepatic steatosis and fibrosis in high-fat diet-fed mice: Role of local renin-angiotensin system, oxidative stress and inflammation. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
42
|
Cao X, Lu XM, Tuo X, Liu JY, Zhang YC, Song LN, Cheng ZQ, Yang JK, Xin Z. Angiotensin-converting enzyme 2 regulates endoplasmic reticulum stress and mitochondrial function to preserve skeletal muscle lipid metabolism. Lipids Health Dis 2019; 18:207. [PMID: 31775868 PMCID: PMC6882339 DOI: 10.1186/s12944-019-1145-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/05/2019] [Indexed: 01/01/2023] Open
Abstract
Objective Endoplasmic reticulum (ER) stress and mitochondrial function affected intramuscular fat accumulation. However, there is no clear evident on the effect of the regulation of ER stress and mitochondrial function by Angiotensin-converting enzyme 2 (ACE2) on the prevention of intramuscular fat metabolism. We investigated the effects of ACE2 on ER stress and mitochondrial function in skeletal muscle lipid metabolism. Methods The triglyceride (TG) content in skeletal muscle of ACE2 knockout mice and Ad-ACE2-treated db/db mice were detected by assay kits. Meanwhile, the expression of lipogenic genes (ACCα, SREBP-1c, LXRα, CPT-1α, PGC-1α and PPARα), ER stress and mitochondrial function related genes (GRP78, eIF2α, ATF4, BCL-2, and SDH6) were analyzed by RT-PCR. Lipid metabolism, ER stress and mitochondrial function related genes were analyzed by RT-PCR in ACE2-overexpression C2C12 cell. Moreover, the IKKβ/NFκB/IRS-1 pathway was determined using lysate sample from skeletal muscle of ACE2 knockout mice. Results ACE2 deficiency in vivo is associated with increased lipid accumulation in skeletal muscle. The ACE2 knockout mice displayed an elevated level of ER stress and mitochondrial dysfunctions in skeletal muscle. In contrast, activation of ACE2 can ameliorate ER stress and mitochondrial function, which slightly accompanied by reduced TG content and down-regulated the expression of skeletal muscle lipogenic proteins in the db/db mice. Additionally, ACE2 improved skeletal muscle lipid metabolism and ER stress genes in the C2C12 cells. Mechanistically, endogenous ACE2 improved lipid metabolism through the IKKβ/NFκB/IRS-1 pathway in skeletal muscle. Conclusions ACE2 was first reported to play a notable role on intramuscular fat regulation by improving endoplasmic reticulum and mitochondrial function. This study may provide a strategy for treating insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
- Xi Cao
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes institute, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xin-Meng Lu
- Department of Endocrinology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Xiu Tuo
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes institute, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jing-Yi Liu
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes institute, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yi-Chen Zhang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes institute, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Li-Ni Song
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes institute, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Zhi-Qiang Cheng
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jin-Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes institute, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Zhong Xin
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes institute, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
43
|
Increased Serum Angiotensin II Is a Risk Factor of Nonalcoholic Fatty Liver Disease: A Prospective Pilot Study. Gastroenterol Res Pract 2019; 2019:5647161. [PMID: 31827504 PMCID: PMC6881577 DOI: 10.1155/2019/5647161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/03/2019] [Indexed: 01/18/2023] Open
Abstract
Background and Aims Nonalcoholic fatty liver disease (NAFLD) is one of the most prevalent chronic liver diseases. In this prospective study, we aim to explore the role of angiotensin II (Ang II) and NLRP3 inflammasome in NAFLD patients. Methods We prospectively enrolled 96 patients in our hospital from September 2014 to February 2016. Patients were divided into two groups (NAFLD group and Control group), and the serum Ang II level, IL-1β, IL-18, and lipids were analyzed. Correlation and multivariable analyses were used in order to identify the potential risk factors of NAFLD. Results Although the two groups share a similar demographic background, the Ang II level of NAFLD group patients was significantly higher than that of the Control group (42.18 ± 12.37 vs. 36.69 ± 13.90, p = 0.014) when abdominal ultrasound was used for grouping. This finding was confirmed when a FibroScan Cap value was selected to divide participants into the NAFLD group and Control group (41.16 ± 13.06 vs. 34.85 ± 12.64, p = 0.040). Multivariable analysis showed that Ang II level is an independent risk factor of NAFLD whether abdominal ultrasound (OR = 1.056, p = 0.037) or FibroScan Cap value (OR = 1.069, p = 0.013) was deemed as the diagnostic standard. Furthermore, stepwise regression analysis was carried out between Ang II with other parameters and we discovered that Ang II had a linear correlation with IL-1β. Conclusion Ang II levels of NAFLD patients significantly increased, and elevated Ang II level is an independent risk factor of NAFLD. Our preliminary results also indicate that Ang II may promote the development of NAFLD by activating NLRP3 inflammasome.
Collapse
|
44
|
Prenatal high-salt diet impaired vasodilatation with reprogrammed renin-angiotensin system in offspring rats. J Hypertens 2019; 36:2369-2379. [PMID: 30382958 DOI: 10.1097/hjh.0000000000001865] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIMS High-salt diet is linked to hypertension, and prenatal high-salt diet increases the risk of cardiovascular diseases in the offspring. The present study investigated whether and how prenatal high-salt diet influenced nitric oxide-mediated vasodilatation in the offspring. METHODS AND RESULTS Pregnant rats were fed either normal-salt (1% sodium chloride) or high-salt (8% sodium chloride) diet during gestation. Experiments were conducted in 5-month-old male offspring. Sodium nitroprusside (SNP, nitric oxide donor)-induced hypotensive responses (in vivo) and vascular dilatation (in vitro) was significantly attenuated (Emax: 84 ± 2 vs. 51 ± 2, high-salt vs. control, P < 0.001) in the high-salt offspring, indicating reduced vascular relaxations. Pretreatment with Tempol (reactive oxygen species scavenger) alleviated this attenuation. The high-salt offspring showed an increased level of oxidative stress markers in both mesenteric arteries and plasma samples. The antioxidant activity, serum superoxide dismutase and catalase were significantly reduced, whereas malondialdehyde was increased in the high-salt offspring. O2 production, and protein expression of Nox2 and Nox4 in mesenteric arteries was significantly increased in the high-salt offspring whereas Nox1 showed no changes. The local renin-angiotensin system in mesenteric arteries was activated, associated with an increased NADPH oxidase. DNA methylation at the proximal promoter of angiotensin-converting enzyme gene in the lung was significantly increased in the high-salt offspring (P = 0.004). CONCLUSION The results suggest that prenatal high-salt diet impairs nitric oxide-mediated vasodilatation because of the increased oxidative stress-affected renin-angiotensin system in the high-salt offspring, providing new information for understanding, and early prevention of cardiovascular diseases in fetal origins.
Collapse
|
45
|
Klein S, Kleine CE, Pieper A, Granzow M, Gautsch S, Himmit M, Kahrmann K, Schierwagen R, Uschner FE, Magdaleno F, Naoum ME, Kristiansen G, Walther T, Bader M, Sauerbruch T, Trebicka J. TGR(mREN2)27 rats develop non-alcoholic fatty liver disease-associated portal hypertension responsive to modulations of Janus-kinase 2 and Mas receptor. Sci Rep 2019; 9:11598. [PMID: 31406138 PMCID: PMC6690919 DOI: 10.1038/s41598-019-48024-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 07/29/2019] [Indexed: 12/14/2022] Open
Abstract
Prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing. Resulting fibrosis and portal hypertension, as a possible secondary event, may necessitate treatment. Overexpression of mouse renin in the transgenic rat model, TGR(mREN2)27, leads to spontaneous development of NAFLD. Therefore, we used TGR(mREN2)27 rats as a model of NAFLD where we hypothesized increased susceptibility and investigated fibrosis and portal hypertension and associated pathways. 12-week old TGR(mREN2)27 rats received either cholestatic (BDL) or toxic injury (CCl4 inhalation). Portal and systemic hemodynamic assessments were performed using microsphere technique with and without injection of the Janus-Kinase 2 (JAK2) inhibitor AG490 or the non-peptidic Ang(1-7) agonist, AVE0991. The extent of liver fibrosis was assessed in TGR(mREN2)27 and wild-type rats using standard techniques. Protein and mRNA levels of profibrotic, renin-angiotensin system components were assessed in liver and primary hepatic stellate cells (HSC) and hepatocytes. TGR(mREN2)27 rats developed spontaneous, but mild fibrosis and portal hypertension due to the activation of the JAK2/Arhgef1/ROCK pathway. AG490 decreased migration of HSC and portal pressure in isolated liver perfusions and in vivo. Fibrosis or portal hypertension after cholestatic (BDL) or toxic injury (CCl4) was not aggravated in TGR(mREN2)27 rats, probably due to decreased mouse renin expression in hepatocytes. Interestingly, portal hypertension was even blunted in TGR(mREN2)27 rats (with or without additional injury) by AVE0991. TGR(mREN2)27 rats are a suitable model of spontaneous liver fibrosis and portal hypertension but not with increased susceptibility to liver damage. After additional injury, the animals can be used to evaluate novel therapeutic strategies targeting Mas.
Collapse
Affiliation(s)
- Sabine Klein
- Department of Internal Medicine I, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Andrea Pieper
- House for Experimental Therapy, University of Bonn, Bonn, Germany
| | - Michaela Granzow
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Sebastian Gautsch
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Mimoun Himmit
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | | | - Robert Schierwagen
- Department of Internal Medicine I, Goethe University Frankfurt, Frankfurt, Germany
| | - Frank Erhard Uschner
- Department of Internal Medicine I, Goethe University Frankfurt, Frankfurt, Germany
| | | | | | | | - Thomas Walther
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland.,Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Bader
- Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Institute for Biology, University of Lübeck, Lübeck, Germany.,Charité-University Medicine Berlin, Berlin, Germany.,Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Tilman Sauerbruch
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, Goethe University Frankfurt, Frankfurt, Germany. .,European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain. .,Institute for Bioengineering of Catalonia, Barcelona, Spain. .,Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
46
|
Cao X, Song LN, Zhang YC, Li Q, Shi TT, Yang FY, Yuan MX, Xin Z, Yang JK. Angiotensin-converting enzyme 2 inhibits endoplasmic reticulum stress-associated pathway to preserve nonalcoholic fatty liver disease. Diabetes Metab Res Rev 2019; 35:e3123. [PMID: 30604460 DOI: 10.1002/dmrr.3123] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/09/2018] [Accepted: 12/23/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND Previous works indicated that the stress on the endoplasmic reticulum (ER) affected nonalcoholic fatty liver disease (NAFLD). However, there is no clear evident on the effect of the regulation of ER stress by angiotensin-converting enzyme 2 (ACE2) on the prevention of NAFLD. METHODS HepG2 cells were treated with thapsigargin (Tg) or palmitic acid (PA). We analysed ACE2 expression using Western-blotting analyses. ER stress-related proteins were detected in ACE2 knockout mice and Ad-ACE2-treated db/db mice by immunofluorescence or Western-blotting analyses. In ACE2-overexpression HepG2 cells, the triglyceride (TG), total cholesterol (TC), and glycogen content were detected by assay kits. Meanwhile, the expression of hepatic lipogenic proteins (ACCα, SREBP-1c, FAS, and LXRα), enzymes for gluconeogenesis (PEPCK, G6Pase, and IRS2), and IKKβ/NFκB/IRS1/Akt pathway were analysed by Western-blotting analyses. RESULTS ACE2 was significantly increased in Tg/PA-induced cultured hepatocytes. Additionally, ACE2 knockout mice displayed elevated levels of ER stress, while Ad-ACE2-treated db/db mice showed reduced ER stress in liver. Furthermore, activation of ACE2 can ameliorate ER stress, accompanied by decreased TG content, increased intracellular glycogen, and downregulated expression of hepatic lipogenic proteins and enzymes for gluconeogenesis in Tg/PA-induced hepatocytes. As a consequence of anti-ER stress, the activation of ACE2 led to improved glucose and lipid metabolism through the IKKβ/NFκB/IRS1/Akt pathway. CONCLUSIONS This is the first time documented that ACE2 had a notable alleviating role in ER stress-induced hepatic steatosis and glucose metabolism via the IKKβ/NFκB/IRS1/Akt-mediated pathway. This study may further provide insight into a novel underlying mechanism and a strategy for treating NAFLD.
Collapse
Affiliation(s)
- Xi Cao
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Diabetes institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Physiology and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Li-Ni Song
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Diabetes institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yi-Chen Zhang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Diabetes institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qi Li
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Diabetes institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ting-Ting Shi
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Diabetes institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Fang-Yuan Yang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Diabetes institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ming-Xia Yuan
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Diabetes institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhong Xin
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Diabetes institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jin-Kui Yang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Diabetes institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
47
|
Sturzeneker MCS, de Noronha L, Olandoski M, Wendling LU, Precoma DB. Ramipril significantly attenuates the development of non-alcoholic steatohepatitis in hyperlipidaemic rabbits. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2019; 9:8-17. [PMID: 31131153 PMCID: PMC6526354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 03/01/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is considered as the most frequent cause of chronic hepatic disease in adults. It is strictly correlated with insulin resistance, frequently associated with components of metabolic syndrome, and, similarly to the latter, it has been correlated with high risk of developing type 2 diabetes and cardiovascular diseases. Systemic arterial hypertension has been suggested to be associated with NAFLD in approximately 40% of the cases, and NAFLD has been independently associated with an increased risk of arterial hypertension in observational studies. Therefore, we can infer that treating arterial hypertension in NAFLD carriers will be often necessary and that the potential beneficial effects of the antihypertensive might, in this context, influence the choice of the respective drug. The renin-angiotensin system has been correlated to the whole basic physiopathogenic mechanism of NAFLD in experimental models. Based on these findings, we conducted this study to evaluate the effects of the ACE-inhibitor ramipril, used preventively, in NAFLD induced in rabbits fed hyperlipidaemic diet. METHODS Twenty-nine rabbits were divided into three groups (normal, placebo, and ramipril). The placebo and ramipril groups were fed a ration containing 0.925% cholesterol. The groups were orally administered 0.35 mg/kg/day of ramipril, and an equivalent volume of vehicle was administered to the placebo group. At the end of the 8th week, all rabbits underwent segmental hepatic resection and were euthanized. Blood samples were collected to determine glucose, insulin, creatinine, total cholesterol, triglycerides, HDL-C, and aminotransferase levels at baseline and euthanasia. Haematoxylin and eosin and Gomori trichrome-stained slides were analysed based on the histological scoring system for NAFLD. Sudan III-stained slides were analysed by morphometry and immunostained based on the Allred scoring system. RESULTS When compared with placebo, ramipril significantly diminished the development of steatosis (P=0.032), lobular inflammation (P=0.006), hepatocellular ballooning (P=0.023), and fibrosis (P=0.02). Based on the NAFLD activity score (NAS), ramipril significantly reduced the development of non-alcoholic steatohepatitis (NASH) (P=0.003). CONCLUSIONS The preventive use of ramipril in rabbits fed hyperlipidaemic diet, attenuates the development of the whole NAFLD histopathological spectrum and based on NAS, ramipril significantly reduced the development of NASH.
Collapse
Affiliation(s)
- Mario Claudio Soares Sturzeneker
- Department of Medicine, Pontificia Universidade Católica do ParanaCuritiba, Brazil
- Department of Medicine, Universidade Estadual de Ponta GrossaPonta Grossa, Brazil
| | - Lucia de Noronha
- Department of Medicine, Pontificia Universidade Católica do ParanaCuritiba, Brazil
| | - Marcia Olandoski
- Department of Medicine, Pontificia Universidade Católica do ParanaCuritiba, Brazil
| | | | | |
Collapse
|
48
|
Borém LMA, Neto JFR, Brandi IV, Lelis DF, Santos SHS. The role of the angiotensin II type I receptor blocker telmisartan in the treatment of non-alcoholic fatty liver disease: a brief review. Hypertens Res 2018; 41:394-405. [PMID: 29636553 PMCID: PMC7091617 DOI: 10.1038/s41440-018-0040-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/31/2017] [Accepted: 11/17/2017] [Indexed: 01/18/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently considered an important component of metabolic syndrome (MetS). The spectrum of NAFLD includes conditions that range from simple hepatic steatosis to non-alcoholic steatohepatitis. NAFLD is correlated with liver-related death and is predicted to be the most frequent indication for liver transplantation by 2030. Insulin resistance is directly correlated to the central mechanisms of hepatic steatosis in NAFLD patients, which is strongly correlated to the imbalance of the renin–angiotensin system, that is involved in lipid and glucose metabolism. Among the emerging treatment approaches for NAFLD is the anti-hypertensive agent telmisartan, which has positive effects on liver, lipid, and glucose metabolism, especially through its action on the renin–angiotensin system, by blocking the ACE/AngII/AT1 axis and increasing ACE2/Ang(1–7)/Mas axis activation. However, treatment with this drug is only recommended for patients with an established indication for anti-hypertensive therapy. Thus, there is an increased need for large randomized controlled trials with the aim of elucidating the effects of telmisartan on liver disease, especially NAFLD. From this perspective, the present review aims to provide a brief examination of the pathogenesis of NAFLD/NASH and the role of telmisartan on preventing liver disorders and thus to improve the discussion on potential therapies.
Collapse
Affiliation(s)
- Luciana M A Borém
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil.,Medicine Department, Faculdades Integradas Pitágoras, Montes Claros, Minas Gerais, Brazil
| | - João F R Neto
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Igor V Brandi
- Institute of Agricultural Sciences, Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Deborah F Lelis
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Sergio H S Santos
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil. .,Institute of Agricultural Sciences, Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil.
| |
Collapse
|
49
|
Gopoju R, Panangipalli S, Kotamraju S. Metformin treatment prevents SREBP2-mediated cholesterol uptake and improves lipid homeostasis during oxidative stress-induced atherosclerosis. Free Radic Biol Med 2018; 118:85-97. [PMID: 29499335 DOI: 10.1016/j.freeradbiomed.2018.02.031] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/06/2018] [Accepted: 02/23/2018] [Indexed: 12/17/2022]
Abstract
Lipids are responsible for the atheromatous plaque formation during atherosclerosis by their deposition in the subendothelial intima of the aorta, leading to infarction. Sterol regulatory element-binding protein 2 (SREBP2), regulating cholesterol homeostasis, is suggested to play a pivotal role during the early incidence of atherosclerosis through dysregulation of lipid homeostasis. Here we demonstrate that oxidative stress stimulates SREBP2-mediated cholesterol uptake via low density lipoprotein receptor (LDLR), rather than cholesterol synthesis, in mouse vascular aortic smooth muscle cells (MOVAS) and THP-1 monocytes. The enhancement of mature form of SREBP2 (SREBP2-M) during oxidative stress was associated with the inhibition of AMP-activated protein kinase (AMPK) activation. In contrast, inhibition of either SREBP2 by fatostatin or LDLR by siLDLR resulted in decreased cholesterol levels during oxidative stress. Thereby confirming the role of SREBP2 in cholesterol regulation via LDLR. Metformin-mediated activation of AMPK was able to significantly abrogate cholesterol uptake by inhibiting SREBP2-M. Interestingly, although metformin administration attenuated angiotensin (Ang)-II-impaired lipid homeostasis in both aorta and liver tissues of ApoE-/- mice, the results indicate that SREBP2 through LDLR regulates lipid homeostasis in aorta but not in liver tissue. Taken together, AMPK activation inhibits oxidative stress-mediated SREBP2-dependent cholesterol uptake, and moreover, metformin-induced prevention of atheromatic events are in part due to its ability to regulate the SREBP2-LDLR axis.
Collapse
Affiliation(s)
- Raja Gopoju
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Training and Development Complex, Chennai 600113, India
| | - Sravya Panangipalli
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, India
| | - Srigiridhar Kotamraju
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Training and Development Complex, Chennai 600113, India.
| |
Collapse
|
50
|
van Breda SG, Claessen SM, van Herwijnen M, Theunissen DH, Jennen DG, de Kok TM, Kleinjans JC. Integrative omics data analyses of repeated dose toxicity of valproic acid in vitro reveal new mechanisms of steatosis induction. Toxicology 2018; 393:160-170. [DOI: 10.1016/j.tox.2017.11.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 10/31/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023]
|