1
|
Nishida N, Sugimoto S, Miyagaki S, Cho C, Konishi M, Goda T, Yamaguchi M, Kawabe Y, Morimoto H, Kusuyama J, Okamura T, Hamaguchi M, Mori J, Nakajima H, Fukui M, Iehara T. Anti-inflammatory effect of Angiotensin 1-7 in white adipose tissue. Adipocyte 2025; 14:2449027. [PMID: 39803918 PMCID: PMC11730366 DOI: 10.1080/21623945.2024.2449027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/08/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025] Open
Abstract
Obesity is a global health concern that promotes chronic low-grade inflammation, leading to insulin resistance, a key factor in many metabolic diseases. Angiotensin 1-7 (Ang 1-7), a component of the renin-angiotensin system (RAS), exhibits anti-inflammatory effects in obesity and related disorders, though its mechanisms remain unclear. In this study, we examined the effect of Ang 1-7 on inflammation of white adipose tissue (WAT) in dietary-induced obese mice. Monocyte chemoattractant protein-1 (MCP-1) produced by white adipocytes and tumour necrosis factor-α (TNF-α) produced by macrophages are pro-inflammatory cytokines and interact to form a pathogenic loop to exacerbate obesity-induced inflammation. We found that Ang 1-7 reduced MCP-1 and TNF-α gene expressions and the number of crown-like structures, which are histological hallmarks of the pro-inflammatory process, in visceral epididymal WAT (eWAT) and reduced circulating MCP-1 and TNF-α levels, accompanied by improvement in insulin resistance, in dietary-induced obese mice. Furthermore, Ang 1-7 reduced MCP-1 and TNF-α secretions in 3T3-L1 white adipocytes and RAW 264.7 macrophages, respectively, which are in vitro experimental models mimicking obesity condition. Our results suggest that Ang 1-7 directly acts on WAT to mitigate obesity-induced inflammation. Thus, this study provides novel insights into the underlying mechanism of anti-obesity effects of Ang 1-7.
Collapse
Affiliation(s)
- Nozomi Nishida
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoru Sugimoto
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Miyagaki
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chiharu Cho
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Madoka Konishi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Goda
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mihoko Yamaguchi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuhiro Kawabe
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hidechika Morimoto
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Joji Kusuyama
- Department of Biosignals and Inheritance, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Takuro Okamura
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jun Mori
- Division of Pediatric Endocrinology, Metabolism and Nephrology, Children’s Medical Center, Osaka City General Hospital, Osaka, Japan
| | - Hisakazu Nakajima
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoko Iehara
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
2
|
Guimarães Júnior OF, Pereira de Oliveira GL, Farias Lelis DD, Faria Baldo TDO, Baldo MP, Sousa Santos SH, Andrade JMO. Expression levels of ACE and ACE2 in the placenta and white adipose tissue of lean and obese pregnant women. Biomarkers 2024; 29:434-441. [PMID: 39348715 DOI: 10.1080/1354750x.2024.2411346] [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: 07/14/2024] [Accepted: 09/22/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND This study evaluated the expression of ACE and ACE2 in the placenta and white adipose tissue in lean and obese women, and correlated their levels with anthropometric, clinical, and laboratory parameters, and tissue count of inflammatory cells. METHODS A cross-sectional analytical study was performed with 49 pregnant women and their respective newborns. Samples of placenta and adipose tissue were used for measuring mRNA expression for ACE and ACE2 through qRT-PCR. Inflammatory cell counting was performed through conventional microscopy. RESULTS An increase in ACE expression and a decrease in ACE2 were observed in the placenta and adipose tissue of women with obesity. ACE2 levels showed a negative correlation with pre-pregnancy BMI and total cholesterol. CONCLUSION Maternal obesity can modulate the expression of RAS components in the placenta and white adipose tissue, with ACE2 correlated with pre-pregnancy BMI and total cholesterol.
Collapse
Affiliation(s)
- Orcione Ferreira Guimarães Júnior
- Graduate Program in Health Sciences (Programa de Pós-Graduação em Ciências da Saúde - PPGCS), State University of Montes Claros (Universidade Estadual de Montes Claros - Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Gabriel Ledo Pereira de Oliveira
- Department of Medicine, Santo Agostinho College - Afya Educacional (Faculdade Santo Agostinho), FASA, Vitória da Conquista, Bahia, Brazil
| | - Deborah de Farias Lelis
- Graduate Program in Health Sciences (Programa de Pós-Graduação em Ciências da Saúde - PPGCS), State University of Montes Claros (Universidade Estadual de Montes Claros - Unimontes), Montes Claros, Minas Gerais, Brazil
- Department of Pathophysiology, Unimontes, Montes Claros, Minas Gerais, Brazil
| | | | - Marcelo Perim Baldo
- Graduate Program in Health Sciences (Programa de Pós-Graduação em Ciências da Saúde - PPGCS), State University of Montes Claros (Universidade Estadual de Montes Claros - Unimontes), Montes Claros, Minas Gerais, Brazil
- Department of Pathophysiology, Unimontes, Montes Claros, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Graduate Program in Health Sciences (Programa de Pós-Graduação em Ciências da Saúde - PPGCS), State University of Montes Claros (Universidade Estadual de Montes Claros - Unimontes), Montes Claros, Minas Gerais, Brazil
- Graduate Program in Food and Health (Programa de Pós-Graduação em Alimentos e Saúde - PPGAS, Federal University of Minas Gerais (Universidade Federal de Minas Gerais - UFMG), Montes Claros, Minas Gerais, Brazil
| | - João Marcus Oliveira Andrade
- Graduate Program in Health Sciences (Programa de Pós-Graduação em Ciências da Saúde - PPGCS), State University of Montes Claros (Universidade Estadual de Montes Claros - Unimontes), Montes Claros, Minas Gerais, Brazil
- Department of Pathophysiology, Unimontes, Montes Claros, Minas Gerais, Brazil
- Graduate Program in Food and Health (Programa de Pós-Graduação em Alimentos e Saúde - PPGAS, Federal University of Minas Gerais (Universidade Federal de Minas Gerais - UFMG), Montes Claros, Minas Gerais, Brazil
| |
Collapse
|
3
|
Akopova O, Korkach Y, Sagach V. The effects of ecdysterone and enalapril on nitric oxide synthesis and the markers of oxidative stress in streptozotocin-induced diabetes in rats: a comparative study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8089-8099. [PMID: 38789633 DOI: 10.1007/s00210-024-03154-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024]
Abstract
Cardiovascular functions in diabetes greatly depend on constitutive NOS (cNOS) activity. A comparative study of the effects of a steroid hormone ecdysterone and enalapril, an ACE inhibitor widely used to treat cardiac disorders on cNOS, inducible NOS (iNOS), xanthine oxidoreductase (XOR) activity, RNS, ROS, and lipid peroxidation in heart tissue in experimental diabetes was conducted. The rat model of diabetes was established by streptozotocin injection. NOS activity, NO2-, NO3-, uric acid, nitrosothiols, hydroperoxide, superoxide, and diene conjugate formation were studied spectrophotomerically. In diabetes, cNOS downregulation correlated with a dramatic fall of NO2- production and ~4.5-fold elevation of nitrosothiols, which agreed with a steep rise of iNOS activity, while NO3- remained close to control. Dramatic activation of XOR was observed, which correlated with the elevation of both superoxide production and nitrate reductase activity and resulted in strong lipid peroxidation. Ecdysterone and enalapril differently affected RNS metabolism. Ecdysterone moderately restored cNOS but strongly suppressed iNOS, which resulted in the reduction of NO3-, but full restoration of NO2- production. Enalapril better restored cNOS but less effectively suppressed iNOS, which promoted NO3- formation. Both drugs similarly inhibited XOR, which equally alleviated oxidative stress and lipid peroxidation. The synergistic action of iNOS and XOR was a plausible explanation for strong lipid peroxidation, abolished by the inhibition of iNOS and XOR by ecdysterone or enalapril. Complementary effects of ecdysterone and enalapril on cNOS, iNOS, and RNS are a promising basis for their combined use in the treatment of cardiovascular disorders caused by cNOS dysfunction in diabetes.
Collapse
Affiliation(s)
- Olga Akopova
- Stem cell laboratory, Bogomoletz Institute of Physiology, NAS of Ukraine, Kiev, Ukraine.
| | - Yulia Korkach
- Circulation department, Bogomoletz Institute of Physiology, NAS of Ukraine, Kiev, Ukraine
| | - Vadim Sagach
- Circulation department, Bogomoletz Institute of Physiology, NAS of Ukraine, Kiev, Ukraine
| |
Collapse
|
4
|
Laurindo LF, Rodrigues VD, Minniti G, de Carvalho ACA, Zutin TLM, DeLiberto LK, Bishayee A, Barbalho SM. Pomegranate (Punica granatum L.) phytochemicals target the components of metabolic syndrome. J Nutr Biochem 2024; 131:109670. [PMID: 38768871 DOI: 10.1016/j.jnutbio.2024.109670] [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: 07/09/2023] [Revised: 04/08/2024] [Accepted: 05/14/2024] [Indexed: 05/22/2024]
Abstract
Pomegranate (Punica granatum L.) is a multipurpose dietary and medicinal plant known for its ability to promote various health benefits. Metabolic syndrome (MetS) is a complex metabolic disorder driving health and socioeconomic challenges worldwide. It may be characterized by insulin resistance, abdominal obesity, hypertension, and dyslipidemia. This study aims to conduct a review of pomegranate's effects on MetS parameters using a mechanistic approach relying on pre-clinical studies. The peel, juice, roots, bark, seeds, flowers, and leaves of the fruit present several bioactive compounds that are related mainly to anti-inflammatory and antioxidant activities as well as cardioprotective, antidiabetic, and antiobesity effects. The use of the juice extract can work as a potent inhibitor of angiotensin-converting enzyme activities, consequently regulating blood pressure. The major bioactive compounds found within the fruit are phenolic compounds (hydrolysable tannins and flavonoids) and fatty acids. Alkaloids, punicalagin, ellagitannins, ellagic acid, anthocyanins, tannins, flavonoids, luteolin, and punicic acid are also present. The antihyperglycemia, antihyperlipidemia, and weight loss promoting effects are likely related to the anti-inflammatory and antioxidant effects. When considering clinical application, pomegranate extracts are found to be frequently well-tolerated, further supporting its efficacy as a treatment modality. We suggest that pomegranate fruit, extract, or processed products can be used to counteract MetS-related risk factors. This review represents an important step towards exploring potential avenues for further research in this area.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), São Paulo, São Paulo, Brazil; Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil
| | - Victória Dogani Rodrigues
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), São Paulo, São Paulo, Brazil
| | - Giulia Minniti
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil
| | - Antonelly Cassio Alves de Carvalho
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil
| | - Tereza Laís Menegucci Zutin
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil
| | - Lindsay K DeLiberto
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL USA
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL USA.
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil; Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), São Paulo, São Paulo, Brazil.
| |
Collapse
|
5
|
Toth DD, Souder CL, Patuel S, English CD, Konig I, Ivantsova E, Malphurs W, Watkins J, Anne Costa K, Bowden JA, Zubcevic J, Martyniuk CJ. Angiotensin II Alters Mitochondrial Membrane Potential and Lipid Metabolism in Rat Colonic Epithelial Cells. Biomolecules 2024; 14:974. [PMID: 39199363 PMCID: PMC11353208 DOI: 10.3390/biom14080974] [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: 07/21/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
An over-active renin-angiotensin system (RAS) is characterized by elevated angiotensin II (Ang II). While Ang II can promote metabolic and mitochondrial dysfunction in tissues, little is known about its role in the gastrointestinal system (GI). Here, we treated rat primary colonic epithelial cells with Ang II (1-5000 nM) to better define their role in the GI. We hypothesized that Ang II would negatively affect mitochondrial bioenergetics as these organelles express Ang II receptors. Ang II increased cellular ATP production but reduced the mitochondrial membrane potential (MMP) of colonocytes. However, cells maintained mitochondrial oxidative phosphorylation and glycolysis with treatment, reflecting metabolic compensation with impaired MMP. To determine whether lipid dysregulation was evident, untargeted lipidomics were conducted. A total of 1949 lipids were detected in colonocytes spanning 55 distinct (sub)classes. Ang II (1 nM) altered the abundance of some sphingosines [So(d16:1)], ceramides [Cer-AP(t18:0/24:0)], and phosphatidylcholines [OxPC(16:0_20:5(2O)], while 100 nM Ang II altered some triglycerides and phosphatidylserines [PS(19:0_22:1). Ang II did not alter the relative expression of several enzymes in lipid metabolism; however, the expression of pyruvate dehydrogenase kinase 2 (PDK2) was increased, and PDK2 can be protective against dyslipidemia. This study is the first to investigate the role of Ang II in colonic epithelial cell metabolism.
Collapse
Affiliation(s)
- Darby D. Toth
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
| | - Christopher L. Souder
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
| | - Sarah Patuel
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
| | - Cole D. English
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
| | - Isaac Konig
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
- Department of Chemistry, Federal University of Lavras (UFLA), Lavras 37200-000, MG, Brazil
| | - Emma Ivantsova
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
| | - Wendi Malphurs
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
| | - Jacqueline Watkins
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
| | - Kaylie Anne Costa
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
| | - John A. Bowden
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
| | - Jasenka Zubcevic
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Block Health Science Bldg, 3000 Arlington Ave, Toledo, OH 43614, USA;
| | - Christopher J. Martyniuk
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (D.D.T.); (C.L.S.II); (S.P.); (C.D.E.); (I.K.); (E.I.); (W.M.); (J.W.); (K.A.C.); (J.A.B.)
- University of Florida Genetics Institute, University of Florida, Gainesville, FL 32611, USA
- Interdisciplinary Program in Biomedical Sciences, Neuroscience, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
6
|
Pan S, A.C. Souza L, Worker CJ, Reyes Mendez ME, Gayban AJB, Cooper SG, Sanchez Solano A, Bergman RN, Stefanovski D, Morton GJ, Schwartz MW, Feng Earley Y. (Pro)renin receptor signaling in hypothalamic tyrosine hydroxylase neurons is required for obesity-associated glucose metabolic impairment. JCI Insight 2024; 9:e174294. [PMID: 38349753 PMCID: PMC11063935 DOI: 10.1172/jci.insight.174294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/08/2024] [Indexed: 03/06/2024] Open
Abstract
Glucose homeostasis is achieved via complex interactions between the endocrine pancreas and other peripheral tissues and glucoregulatory neurocircuits in the brain that remain incompletely defined. Within the brain, neurons in the hypothalamus appear to play a particularly important role. Consistent with this notion, we report evidence that (pro)renin receptor (PRR) signaling within a subset of tyrosine hydroxylase (TH) neurons located in the hypothalamic paraventricular nucleus (PVNTH neurons) is a physiological determinant of the defended blood glucose level. Specifically, we demonstrate that PRR deletion from PVNTH neurons restores normal glucose homeostasis in mice with diet-induced obesity (DIO). Conversely, chemogenetic inhibition of PVNTH neurons mimics the deleterious effect of DIO on glucose. Combined with our finding that PRR activation inhibits PVNTH neurons, these findings suggest that, in mice, (a) PVNTH neurons play a physiological role in glucose homeostasis, (b) PRR activation impairs glucose homeostasis by inhibiting these neurons, and (c) this mechanism plays a causal role in obesity-associated metabolic impairment.
Collapse
Affiliation(s)
- Shiyue Pan
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Lucas A.C. Souza
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Caleb J. Worker
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Miriam E. Reyes Mendez
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Ariana Julia B. Gayban
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Silvana G. Cooper
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Alfredo Sanchez Solano
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Darko Stefanovski
- New Bolton Center, School of Veterinary Medicine, University of Pennsylvania Philadelphia, Pennsylvania, USA
| | - Gregory J. Morton
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, Washington, USA
| | - Michael W. Schwartz
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, Washington, USA
| | - Yumei Feng Earley
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| |
Collapse
|
7
|
Imenshahidi M, Roohbakhsh A, Hosseinzadeh H. Effects of telmisartan on metabolic syndrome components: a comprehensive review. Biomed Pharmacother 2024; 171:116169. [PMID: 38228033 DOI: 10.1016/j.biopha.2024.116169] [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: 11/19/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 01/18/2024] Open
Abstract
Telmisartan is an antagonist of the angiotensin II receptor used in the management of hypertension (alone or in combination with other antihypertensive agents. It belongs to the drug class of angiotensin II receptor blockers (ARBs). Among drugs of this class, telmisartan shows particular pharmacologic properties, including a longer half-life than any other angiotensin II receptor blockers that bring higher and persistent antihypertensive activity. In hypertensive patients, telmisartan has superior efficacy than other antihypertensive drugs (losartan, valsartan, ramipril, atenolol, and perindopril) in controlling blood pressure, especially towards the end of the dosing interval. Telmisartan has a partial PPARγ-agonistic effect whilst does not have the safety concerns of full agonists of PPARγ receptors (thiazolidinediones). Moreover, telmisartan has an agonist activity on PPARα and PPARδ receptors and modulates the adipokine levels. Thus, telmisartan could be considered as a suitable alternative option, with multi-benefit for all components of metabolic syndrome including hypertension, diabetes mellitus, obesity, and hyperlipidemia. This review will highlight the role of telmisartan in metabolic syndrome and the main mechanisms of action of telmisartan are discussed and summarized. Many studies have demonstrated the useful properties of telmisartan in the prevention and improving of metabolic syndrome and this well-tolerated drug can be greatly proposed in the treatment of different components of metabolic syndrome. However, larger and long-duration studies are needed to confirm these findings in long-term observational studies and prospective trials and to determine the optimum dose of telmisartan in metabolic syndrome.
Collapse
Affiliation(s)
- Mohsen Imenshahidi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Arena G, Kitsos A, Hamdorf JM, D’Arcy‐Evans M, Kilpatrick M, Venn A, Preen DB. Evaluation of prescription medication changes following sleeve gastrectomy surgery. Obes Sci Pract 2024; 10:e742. [PMID: 38352066 PMCID: PMC10863745 DOI: 10.1002/osp4.742] [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: 10/06/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Objective The increasing global prevalence of obesity, coupled with its association with chronic health conditions and rising healthcare costs, highlights the need for effective interventions; however, despite the availability of treatment options, the ongoing success of primary interventions in maintaining long-term weight loss remains limited. This study examined the prescription medication dispensing changes following sleeve gastrectomy in Australians aged 45 years and over. Methods In a retrospective analysis of 847 bariatric surgery patients from the New South Wales 45 and Up Study, the assessment of medication patterns categorizing into three groups: gastrointestinal, metabolic, cardiorespiratory, musculoskeletal, and nervous systems was conducted. Each drug class was analyzed, focusing on patients with dispensing records within the 12 months before surgery. This study employed interrupted time-series analysis to compare pre- and post-surgery medication usage. Results With a predominantly female population (76.9%) and an average age of 57.2 (standard deviation 5.71), there were statistically significant reductions in both unique medications (12.5% decrease, p = 0.004) and total medications dispensed (15.9% decrease, p = 0.003) from 12 months before surgery to 13-24 months after bariatric surgery. All medication categories, except opioids, showed reductions. Notably, the most significant reductions were observed in diabetes (38.6%), agents acting on the renin-angiotensin system (40.4%), lipid modifying agents (26.5%), anti-inflammatory products (46.3%), and obstructive airway diseases (53.3%) medications during this time frame. Conclusion These findings suggest that sleeve gastrectomy provides an effective therapeutic intervention for patients with comorbidities requiring multiple medications, especially for obesity-related diseases such as diabetes, cardiovascular, respiratory and musculoskeletal disorders.
Collapse
Affiliation(s)
- Gina Arena
- School of Population and Global HealthThe University of Western AustraliaNedlandsWestern AustraliaAustralia
| | - Alex Kitsos
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - Jeffrey M. Hamdorf
- Medical SchoolThe University of Western AustraliaNedlandsWestern AustraliaAustralia
- Western Surgical HealthNedlandsWestern AustraliaAustralia
| | | | - Michelle Kilpatrick
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - Alison Venn
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - David B. Preen
- School of Population and Global HealthThe University of Western AustraliaNedlandsWestern AustraliaAustralia
| |
Collapse
|
9
|
Chaimati S, Shantavasinkul PC, Sritara P, Sirivarasai J. Effects of AGT and AGTR1 Genetic Polymorphisms and Changes in Blood Pressure Over a Five-Year Follow-Up. Risk Manag Healthc Policy 2023; 16:2931-2942. [PMID: 38164294 PMCID: PMC10758254 DOI: 10.2147/rmhp.s442983] [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: 10/18/2023] [Accepted: 12/15/2023] [Indexed: 01/03/2024] Open
Abstract
Purpose The renin-angiotensin system plays an important role in the central regulation of blood pressure (BP). Genetic variations of angiotensinogen (AGT) and angiotensin II type 1 receptor (AGTR1) may increase susceptibility to elevated BP and hypertension. This study investigated the effects of AGT rs699 and AGTR1 rs5186 single nucleotide polymorphisms (SNPs) on BP at baseline and at a 5-year follow-up. Paticipants and Methods The study population consisted of participants from the Electricity Generating Authority of Thailand cohort study (n=354); data were collected at baseline (2013) and 5 years later (2018). Genotyping of the two SNPs was performed using TaqMan® assay and statistical analyses were performed with SNPStats software. Results The frequencies of the two SNPs were within the Hardy-Weinberg equilibrium (p=0.22 for AGT rs699 and p=0.06 for AGTR1 rs5186). For each SNP, mutant genotypes were significantly associated with increased systolic BP and/or diastolic BP in the codominant and recessive models. Risk alleles of AGT rs699 and AGTR1 rs5186 were associated with increased odds of hypertension and hypertension with metabolic syndrome at follow-up. Conclusion Overall, our results suggest that polymorphisms of genes in the renin-angiotensin system increase susceptibility to the development and progression of hypertension and the development of the metabolic syndrome.
Collapse
Affiliation(s)
- Siwaphorn Chaimati
- Doctoral Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of Nutrition, Mahidol University, Bangkok, 10400, Thailand
| | | | - Piyamitr Sritara
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Jintana Sirivarasai
- Nutrition Division, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
10
|
Guimarães JPT, Queiroz LAD, Menikdiwela KR, Pereira N, Ramalho T, Jancar S, Moustaid-Moussa N, Martins JO. The role of captopril in leukotriene deficient type 1 diabetic mice. Sci Rep 2023; 13:22105. [PMID: 38092813 PMCID: PMC10719306 DOI: 10.1038/s41598-023-49449-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
T1D can be associated with metabolic disorders and several impaired pathways, including insulin signaling, and development of insulin resistance through the renin-angiotensin system (RAS). The main precursor of RAS is angiotensinogen (Agt) and this system is often linked to autophagy dysregulation. Dysregulated autophagy has been described in T1D and linked to impairments in both glucose metabolism, and leukotrienes (LTs) production. Here, we have investigated the role of RAS and LTs in both muscle and liver from T1D mice, and its effects on insulin and autophagy pathways. We have chemically induced T1D in 129sve and 129sve 5LO-/- mice (lacking LTs) with streptozotocin (STZ). To further inhibit ACE activity, mice were treated with captopril (Cap). In muscle of T1D mice, treatment with Cap increased the expression of RAS (angiotensinogen and angiotensin II receptor), insulin signaling, and autophagy markers, regardless of the genotype. In the liver of T1D mice, the treatment with Cap increased the expression of RAS and insulin signaling markers, mostly when LTs were absent. 5LO-/- T1D mice showed increased insulin sensitivity, and decreased NEFA, after the Cap treatment. Cap treatment impacted both insulin signaling and autophagy pathways at the mRNA levels in muscle and liver, indicating the potential role of ACE inhibition on insulin sensitivity and autophagy in T1D.
Collapse
Affiliation(s)
- João Pedro Tôrres Guimarães
- Laboratory of Immunoendocrinology, School of Pharmaceutical Sciences, Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, SP, Brazil
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, and Obesity Research Institute, Texas Tech University (TTU), Lubbock, TX, USA
- Laboratory of Immunopharmacology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo, SP, Brazil
| | - Luiz A D Queiroz
- Laboratory of Immunoendocrinology, School of Pharmaceutical Sciences, Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, SP, Brazil
| | - Kalhara R Menikdiwela
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, and Obesity Research Institute, Texas Tech University (TTU), Lubbock, TX, USA
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Nayara Pereira
- Laboratory of Immunopharmacology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo, SP, Brazil
- Department of Pharmacology, Ribeirão Preto Medical School (FMRP/USP), Ribeirão Preto, SP, Brazil
| | - Theresa Ramalho
- Laboratory of Immunopharmacology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo, SP, Brazil
- Department of Molecular Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sonia Jancar
- Laboratory of Immunopharmacology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo, SP, Brazil
| | - Naima Moustaid-Moussa
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, and Obesity Research Institute, Texas Tech University (TTU), Lubbock, TX, USA.
| | - Joilson O Martins
- Laboratory of Immunoendocrinology, School of Pharmaceutical Sciences, Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
11
|
Pedrosa MA, Labandeira CM, Lago-Baameiro N, Valenzuela R, Pardo M, Labandeira-Garcia JL, Rodriguez-Perez AI. Extracellular Vesicles and Their Renin-Angiotensin Cargo as a Link between Metabolic Syndrome and Parkinson's Disease. Antioxidants (Basel) 2023; 12:2045. [PMID: 38136165 PMCID: PMC10741149 DOI: 10.3390/antiox12122045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Several studies showed an association between metabolic syndrome (MetS) and Parkinson's disease (PD). The linking mechanisms remain unclear. MetS promotes low-grade peripheral oxidative stress and inflammation and dysregulation of the adipose renin-angiotensin system (RAS). Interestingly, brain RAS dysregulation is involved in the progression of dopaminergic degeneration and PD. Circulating extracellular vesicles (EVs) from MetS fat tissue can cross the brain-blood barrier and may act as linking signals. We isolated and characterized EVs from MetS and control rats and analyzed their mRNA and protein cargo using RT-PCR and the ExoView R200 platform, respectively. Furthermore, cultures of the N27 dopaminergic cell line and the C6 astrocytic cell line were treated with EVs from MetS rats. EVs were highly increased in MetS rat serum, which was inhibited by treatment of the rats with the angiotensin type-1-receptor blocker candesartan. Furthermore, EVs from MetS rats showed increased pro-oxidative/pro-inflammatory and decreased anti-oxidative/anti-inflammatory RAS components, which were inhibited in candesartan-treated MetS rats. In cultures, EVs from MetS rats increased N27 cell death and modulated C6 cell function, upregulating markers of neuroinflammation and oxidative stress, which were inhibited by the pre-treatment of cultures with candesartan. The results from rat models suggest EVs and their RAS cargo as a mechanism linking Mets and PD.
Collapse
Affiliation(s)
- Maria A. Pedrosa
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | | | - Nerea Lago-Baameiro
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, 15706 Santiago de Compostela, Spain; (N.L.-B.); (M.P.)
| | - Rita Valenzuela
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Maria Pardo
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, 15706 Santiago de Compostela, Spain; (N.L.-B.); (M.P.)
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jose Luis Labandeira-Garcia
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Ana I. Rodriguez-Perez
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
12
|
Alla JA, Nerger E, Langer A, Quitterer U. Identification of membrane palmitoylated protein 1 (MPP1) as a heart-failure-promoting protein triggered by cardiovascular risk factors and aging. Biochem Pharmacol 2023; 217:115789. [PMID: 37683843 DOI: 10.1016/j.bcp.2023.115789] [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: 07/14/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
Membrane-Associated Guanylate Kinase (MAGUK) proteins are scaffold proteins with well-established functions in the neuronal system. A role of MAGUK protein up-regulation in the pathogenesis of heart failure is not established. This study identified the up-regulation of the MAGUK family protein MPP1 (Membrane Palmitoylated Protein 1), in cardiac transcriptome data of three different heart failure models. MPP1 was up-regulated in failing hearts of B6 mice with long-term chronic pressure overload, in failing hearts of aged Apoe-/- mice with long-term atherosclerosis, and in failing hearts of RKIP-transgenic mice with cardiotoxic lipid overload. MPP1-transgenic mice revealed that moderately (2-fold) increased cardiac MPP1 levels caused symptoms of heart failure with a significantly reduced left ventricular ejection fraction of 39.0 ± 6.9 % in Tg-MPP1 mice compared to 55.2 ± 3.7 % of non-transgenic B6 controls. Echocardiographic and histological analyses detected cardiac enlargement and cardiac dilation in Tg-MPP1 mice. The angiotensin II AT1 receptor (AGTR1) and MPP1 were co-localized on sarcolemmal membranes in vivo, and Tg-MPP1 mice had increased levels of cardiac AGTR1, which has an established heart failure-promoting function. The increased AGTR1 protein could be directly triggered by elevated MPP1 because MPP1 also increased the AGTR1 protein in non-cardiomyocyte HEK cells, which was detected by fluorescence measurement of AGTR1eYFP. MPP1 was not only up-regulated by major cardiovascular risk factors but also by old age, which is a major contributor to heart failure. Thus, the aging-induced MPP1 exerts a previously unrecognized role in heart failure pathogenesis by upregulation of the angiotensin II AT1 receptor (AGTR1) protein.
Collapse
Affiliation(s)
- Joshua Abd Alla
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | - Eric Nerger
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Andreas Langer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Ursula Quitterer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
| |
Collapse
|
13
|
Mohater S, Qahtan S, Alrefaie Z, Alahmadi A. Vitamin D improves hepatic alterations in ACE1 and ACE2 expression in experimentally induced metabolic syndrome. Saudi Pharm J 2023; 31:101709. [PMID: 37559868 PMCID: PMC10407910 DOI: 10.1016/j.jsps.2023.101709] [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: 04/15/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023] Open
Abstract
Metabolic Syndrome (MetS) is a term used to describe a cluster of pathophysiological, biochemical, and metabolic criteria; including high Blood Pressure (BP), high cholesterol, dyslipidaemia, central obesity and Insulin Resistance (IR). The Renin Angiotensin System (RAS) has a regulatory function in BP, hydroelectrolyte balance, and cardiovascular function. RAS is composed of angiotensinogen (AGT), (Ang I), (Ang II), (ACE1), (ACE2), (AT1R), (AT2R), and (Ang 1-7). Vitamin D had been proved to act as a protective factor against MetS. Therefore, the study is pursued to explore vitamin D supplementation roles on hepatic RAS in MetS experimental model. At first, 36 males Albino rats were separated into 4 groups and induced to MetS under controlled circumstances for 3 months. Then, data were collected from blood samples, whereas RNA extracted from liver were analyzed using biochemical and statistical analysis tests. As a result, the major finding was proving that vitamin D can balance the expression of ACE1 and ACE2. Also, confirming that it can improve MetS components by elevating HDL and insulin levels while reducing the levels of BP, cholesterol, LDL, TG, GLU, ALT, AST, and IR. These outcomes may give a new insight into the RAS pathways associated with MetS.
Collapse
Affiliation(s)
- Sara Mohater
- Department of Biological Sciences, College of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Samar Qahtan
- Department of Biological Sciences, College of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zienab Alrefaie
- Medical Physiology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Physiology Department, Faculty of Medicine, Cairo University, Egypt
| | - Ahlam Alahmadi
- Department of Biological Sciences, College of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
14
|
Pan S, Worker CJ, Feng Earley Y. The hypothalamus as a key regulator of glucose homeostasis: emerging roles of the brain renin-angiotensin system. Am J Physiol Cell Physiol 2023; 325:C141-C154. [PMID: 37273237 PMCID: PMC10312332 DOI: 10.1152/ajpcell.00533.2022] [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: 11/29/2022] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/06/2023]
Abstract
The regulation of plasma glucose levels is a complex and multifactorial process involving a network of receptors and signaling pathways across numerous organs that act in concert to ensure homeostasis. However, much about the mechanisms and pathways by which the brain regulates glycemic homeostasis remains poorly understood. Understanding the precise mechanisms and circuits employed by the central nervous system to control glucose is critical to resolving the diabetes epidemic. The hypothalamus, a key integrative center within the central nervous system, has recently emerged as a critical site in the regulation of glucose homeostasis. Here, we review the current understanding of the role of the hypothalamus in regulating glucose homeostasis, with an emphasis on the paraventricular nucleus, the arcuate nucleus, the ventromedial hypothalamus, and lateral hypothalamus. In particular, we highlight the emerging role of the brain renin-angiotensin system in the hypothalamus in regulating energy expenditure and metabolic rate, as well as its potential importance in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- Shiyue Pan
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Caleb J Worker
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Yumei Feng Earley
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| |
Collapse
|
15
|
Nogueiras R, Aspichueta P. Expanding the roles of the renin-angiotensin system: Drug-induced liver injury. J Hepatol 2023; 78:457-459. [PMID: 36592643 DOI: 10.1016/j.jhep.2022.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022]
Affiliation(s)
- Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain; Galician Agency of Investigation, Xunta de Galicia, Spain.
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Spain; CIBER Enfermedades Hepáticas y Digestivas (CIBERehd), Spain.
| |
Collapse
|
16
|
Borkotoky S, Dey D, Hazarika Z. Interactions of angiotensin-converting enzyme-2 (ACE2) and SARS-CoV-2 spike receptor-binding domain (RBD): a structural perspective. Mol Biol Rep 2023; 50:2713-2721. [PMID: 36562937 PMCID: PMC9786537 DOI: 10.1007/s11033-022-08193-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has caused millions of infections and deaths worldwide since its discovery in late 2019 in Wuhan, China. The receptor-binding domain (RBD) of the SARS-CoV-2 spike protein binds to the human angiotensin-converting enzyme-2 (ACE2) receptor, a critical component of the renin-angiotensin system (RAS) that initiates the viral transmission. Most of the critical mutations found in SARS-CoV-2 are associated with the RBD of the spike protein. These mutations have the potential to reduce the efficacy of vaccines and neutralizing antibodies. METHODS In this review, the structural details of ACE2, RBD and their interactions are discussed. In addition, some critical mutations of RBD and their impact on ACE2-RBD interactions are also discussed. CONCLUSION Preventing the interaction between Spike RBD and ACE2 is considered a viable therapeutic strategy since ACE2 binding by RBD is the first step in virus infection. Because the interactions between the two entities are critical for both viral transmission and therapeutic development, it is essential to understand their interactions in detail.
Collapse
Affiliation(s)
- Subhomoi Borkotoky
- Department of Biotechnology, Faculty of Biosciences, Invertis University, Bareilly, Uttar Pradesh, 243123, India.
| | - Debajit Dey
- School of Medicine, University of Maryland Baltimore, Baltimore, MD, 21201, United States of America
| | | |
Collapse
|
17
|
Borborema MEDA, de Lucena TMC, Silva JDA. Vitamin D and estrogen steroid hormones and their immunogenetic roles in Infectious respiratory (TB and COVID-19) diseases. Genet Mol Biol 2023; 46:e20220158. [PMID: 36745756 PMCID: PMC9901533 DOI: 10.1590/1415-4757-gmb-2022-0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 12/07/2022] [Indexed: 02/08/2023] Open
Abstract
The role of steroid hormones against infectious diseases has been extensively studied. From immunomodulatory action to direct inhibition of microorganism growth, hormones D3 (VD3) and 17β-estradiol (E2), and the genetic pathways modulated by them, are key targets for a better understanding pathogenesis of infectious respiratory diseases (IRD) such as tuberculosis (TB) and the coronavirus disease-19 (COVID-19). Currently, the world faces two major public health problems, the outbreak of COVID-19, accounting for more than 6 million so far, and TB, more than 1 million deaths per year. Both, although resulting from different pathogens, the Mtb and the SARS-CoV-2, respectively, are considered serious and epidemic. TB and COVID-19 present similar infection rates between men and women, however the number of complications and deaths resulting from the two infections is higher in men when compared to women in childbearing age, which may indicate a role of the sex hormone E2 in the context of these diseases. E2 and VD3 act upon key gene pathways as important immunomodulatory players and supporting molecules in IRDs. This review summarizes the main roles of these hormones (VD3 and E2) in modulating immune and inflammatory responses and their relationship with TB and COVID-19.
Collapse
Affiliation(s)
- Maria Eduarda de Albuquerque Borborema
- Universidade Federal de Pernambuco, Departamento de Genética, Laboratório de Genética e Biologia Molecular Humana (LGBMH), Recife, PE, Brazil
- Universidade Federal de Pernambuco, Laboratório de Imunopatologia Keizo Asami (LIKA), Recife, PE, Brazil
| | - Thays Maria Costa de Lucena
- Universidade Federal de Pernambuco, Departamento de Genética, Laboratório de Genética e Biologia Molecular Humana (LGBMH), Recife, PE, Brazil
- Universidade Federal de Pernambuco, Laboratório de Imunopatologia Keizo Asami (LIKA), Recife, PE, Brazil
| | - Jaqueline de Azevêdo Silva
- Universidade Federal de Pernambuco, Departamento de Genética, Laboratório de Genética e Biologia Molecular Humana (LGBMH), Recife, PE, Brazil
- Universidade Federal de Pernambuco, Laboratório de Imunopatologia Keizo Asami (LIKA), Recife, PE, Brazil
| |
Collapse
|
18
|
AT1 receptor autoantibodies mediate effects of metabolic syndrome on dopaminergic vulnerability. Brain Behav Immun 2023; 108:255-268. [PMID: 36535607 DOI: 10.1016/j.bbi.2022.12.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/20/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The metabolic syndrome has been associated to chronic peripheral inflammation and related with neuroinflammation and neurodegeneration, including Parkinson's disease. However, the responsible mechanisms are unclear. Previous studies have involved the brain renin-angiotensin system in progression of Parkinson's disease and the angiotensin receptor type 1 (AT1) has been recently revealed as a major marker of dopaminergic vulnerability in humans. Dysregulation of tissue renin-angiotensin system is a key common mechanism for all major components of metabolic syndrome. Circulating AT1 agonistic autoantibodies have been observed in several inflammation-related peripheral processes, and activation of AT1 receptors of endothelial cells, dopaminergic neurons and glial cells have been observed to disrupt endothelial blood -brain barrier and induce neurodegeneration, respectively. Using a rat model, we observed that metabolic syndrome induces overactivity of nigral pro-inflammatory renin-angiotensin system axis, leading to increase in oxidative stress and neuroinflammation and enhancing dopaminergic neurodegeneration, which was inhibited by treatment with AT1 receptor blockers (ARBs). In rats, metabolic syndrome induced the increase in circulating levels of LIGHT and other major pro-inflammatory cytokines, and 27-hydroxycholesterol. Furthermore, the rats showed a significant increase in serum levels of proinflammatory AT1 and angiotensin converting enzyme 2 (ACE2) autoantibodies, which correlated with levels of several metabolic syndrome parameters. We also found AT1 and ACE2 autoantibodies in the CSF of these rats. Effects of circulating autoantibodies were confirmed by chronic infusion of AT1 autoantibodies, which induced blood-brain barrier disruption, an increase in the pro-inflammatory renin-angiotensin system activity in the substantia nigra and a significant enhancement in dopaminergic neuron death in two different rat models of Parkinson's disease. Observations in the rat models, were analyzed in a cohort of parkinsonian and non-parkinsonian patients with or without metabolic syndrome. Non-parkinsonian patients with metabolic syndrome showed significantly higher levels of AT1 autoantibodies than non-parkinsonian patients without metabolic syndrome. However, there was no significant difference between parkinsonian patients with metabolic syndrome or without metabolic syndrome, which showed higher levels of AT1 autoantibodies than non-parkinsonian controls. This is consistent with our recent studies, showing significant increase of AT1 and ACE2 autoantibodies in parkinsonian patients, which was related to dopaminergic degeneration and neuroinflammation. Altogether may lead to a vicious circle enhancing the progression of the disease that may be inhibited by strategies against production of these autoantibodies or AT1 receptor blockers (ARBs).
Collapse
|
19
|
Badr AM, Sherif IO, Mahran YF, Attia HA. Role of Renin-Angiotensin System in the Pathogenesis and Progression of Non-alcoholic Fatty Liver. THE RENIN ANGIOTENSIN SYSTEM IN CANCER, LUNG, LIVER AND INFECTIOUS DISEASES 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]
|
20
|
Giotis ES, Cil E, Brooke GN. Use of Antiandrogens as Therapeutic Agents in COVID-19 Patients. Viruses 2022; 14:2728. [PMID: 36560732 PMCID: PMC9788624 DOI: 10.3390/v14122728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
COVID-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS CoV-2), is estimated to have caused over 6.5 million deaths worldwide. The emergence of fast-evolving SARS-CoV-2 variants of concern alongside increased transmissibility and/or virulence, as well as immune and vaccine escape capabilities, highlight the urgent need for more effective antivirals to combat the disease in the long run along with regularly updated vaccine boosters. One of the early risk factors identified during the COVID-19 pandemic was that men are more likely to become infected by the virus, more likely to develop severe disease and exhibit a higher likelihood of hospitalisation and mortality rates compared to women. An association exists between SARS-CoV-2 infectiveness and disease severity with sex steroid hormones and, in particular, androgens. Several studies underlined the importance of the androgen-mediated regulation of the host protease TMPRSS2 and the cell entry protein ACE2, as well as the key role of these factors in the entry of the virus into target cells. In this context, modulating androgen signalling is a promising strategy to block viral infection, and antiandrogens could be used as a preventative measure at the pre- or early hospitalisation stage of COVID-19 disease. Different antiandrogens, including commercial drugs used to treat metastatic castration-sensitive prostate cancer and other conditions, have been tested as antivirals with varying success. In this review, we summarise the most recent updates concerning the use of antiandrogens as prophylactic and therapeutic options for COVID-19.
Collapse
Affiliation(s)
- Efstathios S. Giotis
- Department of Infectious Diseases, Imperial College London, London W2 1PG, UK
- School of Life Sciences, University of Essex, Colchester CO4 3SQ, UK
| | - Emine Cil
- School of Life Sciences, University of Essex, Colchester CO4 3SQ, UK
| | - Greg N. Brooke
- School of Life Sciences, University of Essex, Colchester CO4 3SQ, UK
| |
Collapse
|
21
|
Devaux CA, Camoin-Jau L. An update on angiotensin-converting enzyme 2 structure/functions, polymorphism, and duplicitous nature in the pathophysiology of coronavirus disease 2019: Implications for vascular and coagulation disease associated with severe acute respiratory syndrome coronavirus infection. Front Microbiol 2022; 13:1042200. [PMID: 36519165 PMCID: PMC9742611 DOI: 10.3389/fmicb.2022.1042200] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/07/2022] [Indexed: 08/01/2023] Open
Abstract
It has been known for many years that the angiotensin-converting enzyme 2 (ACE2) is a cell surface enzyme involved in the regulation of blood pressure. More recently, it was proven that the severe acute respiratory syndrome coronavirus (SARS-CoV-2) interacts with ACE2 to enter susceptible human cells. This functional duality of ACE2 tends to explain why this molecule plays such an important role in the clinical manifestations of coronavirus disease 2019 (COVID-19). At the very start of the pandemic, a publication from our Institute (entitled "ACE2 receptor polymorphism: susceptibility to SARS-CoV-2, hypertension, multi-organ failure, and COVID-19 disease outcome"), was one of the first reviews linking COVID-19 to the duplicitous nature of ACE2. However, even given that COVID-19 pathophysiology may be driven by an imbalance in the renin-angiotensin system (RAS), we were still far from understanding the complexity of the mechanisms which are controlled by ACE2 in different cell types. To gain insight into the physiopathology of SARS-CoV-2 infection, it is essential to consider the polymorphism and expression levels of the ACE2 gene (including its alternative isoforms). Over the past 2 years, an impressive amount of new results have come to shed light on the role of ACE2 in the pathophysiology of COVID-19, requiring us to update our analysis. Genetic linkage studies have been reported that highlight a relationship between ACE2 genetic variants and the risk of developing hypertension. Currently, many research efforts are being undertaken to understand the links between ACE2 polymorphism and the severity of COVID-19. In this review, we update the state of knowledge on the polymorphism of ACE2 and its consequences on the susceptibility of individuals to SARS-CoV-2. We also discuss the link between the increase of angiotensin II levels among SARS-CoV-2-infected patients and the development of a cytokine storm associated microvascular injury and obstructive thrombo-inflammatory syndrome, which represent the primary causes of severe forms of COVID-19 and lethality. Finally, we summarize the therapeutic strategies aimed at preventing the severe forms of COVID-19 that target ACE2. Changing paradigms may help improve patients' therapy.
Collapse
Affiliation(s)
- Christian A. Devaux
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU–Méditerranée Infection, Marseille, France
- Center National de la Recherche Scientifique, Marseille, France
| | - Laurence Camoin-Jau
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU–Méditerranée Infection, Marseille, France
- Laboratoire d’Hématologie, Hôpital de La Timone, APHM, Boulevard Jean-Moulin, Marseille, France
| |
Collapse
|
22
|
Effects of captopril on glucose metabolism and autophagy in liver and muscle from mice with type 1 diabetes and diet-induced obesity. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166477. [PMID: 35780942 DOI: 10.1016/j.bbadis.2022.166477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022]
Abstract
Impaired metabolic functions underlie the pathophysiology of diabetes and obesity. The renin-angiotensin system (RAS) is one pathway related to the pathophysiology of both diseases. RAS activation in metabolically active tissues exerts pro-inflammatory effects via angiotensin II (Ang II), linked to dysfunction in cellular processes such as autophagy, which is associated with obesity and diabetes. Here, we determined whether RAS is involved in metabolic dysregulations in a Type 1 Diabetes (T1D) mouse model, treated with captopril, and in an obesity mouse model (Agt-Tg) that overexpresses angiotensinogen (Agt) in adipose tissue. T1D mice had lower plasma leptin, resistin and higher non-esterified fatty acids (NEFA) compared to wild type (Wt) mice, even under captopril treatment. Further, mRNA levels for Agt, At1, Insr, and Beclin1 were upregulated in muscle and liver of T1D mice with captopril compared to Wt. Moreover, autophagy markers LC3 and p62 proteins were decreased, regardless of captopril treatment in the liver from T1D mice. In obese Wt mice, captopril increased muscle Irs1 gene levels. Further, captopril reduced mRNA levels of At1, Insr, Ampk, Beclin1, Atg12, and Lc3 in the liver from both Wt and Agt-Tg mice, while Agt, At1, Insr, and Atg12 expression was reduced in Agt-Tg mice without captopril treatment. Irs1 expression was decreased in the liver from obese Wt mice treated with captopril. Our results suggest that captopril treatment upregulates components of RAS, insulin signaling, and autophagy in both muscle and liver, indicating potential utility of captopril in targeting both insulin sensitivity and autophagy in diabetes and obesity.
Collapse
|
23
|
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: 4] [Impact Index Per Article: 1.3] [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
|
24
|
Khaksari M, Raji-Amirhasani A, Bashiri H, Ebrahimi MN, Azizian H. Protective effects of combining SERMs with estrogen on metabolic parameters in postmenopausal diabetic cardiovascular dysfunction: The role of cytokines and angiotensin II. Steroids 2022; 183:109023. [PMID: 35358567 DOI: 10.1016/j.steroids.2022.109023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 03/23/2022] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The beneficial effects of the administration of selective estrogen receptor modulators (SERMs) and estrogen (E2), alone or in combination with each other, have been reported in postmenopausal diabetic cardiovascular dysfunction. In the present study, we determined the mechanism of action of SERMs and E2 on inflammatory balance, angiotensin II (Ang II) serum levels, and glycemic profile in a postmenopausal diabetic rat model. METHODS Ovariectomized rats with type 2 diabetes received daily SERMs (tamoxifen and raloxifene) and E2 for one month. After treatment, cardiovascular risk indices, glycemic profile, and serum Ang II, TNF-α and IL-10 levels were measured. RESULTS Type 2 diabetes caused an abnormal glycemic profile, which was exacerbated by ovariectomy. All treatments inhibited the effects of diabetes and ovariectomy on the glycemic profile, with combined treatments (SERMs + E2) showing stronger effects. Cardiovascular risk indices that became abnormal by diabetes and worsened by ovariectomy were improved in all treatment modalities. Also, combined treatment reduced serum Ang II, TNF-α, and the ratio of TNF-α to IL-10, indicating an improvement in inflammatory balance. CONCLUSION Our study showed the administration of SERMs and E2, alone or in combination, could be an effective alternative in the treatment of menopausal diabetes, and generally, the beneficial effects of combined treatments were more effective than the effects of E2 or SERMs alone. It appears that E2 or SERMs benefit the cardiovascular system by improving inflammatory balance and reducing Ang II levels.
Collapse
Affiliation(s)
- Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Alireza Raji-Amirhasani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Cardiovascular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Hossein Azizian
- Neurobiomedical Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
25
|
Sengupta P, Dutta S. N-acetyl cysteine as a potential regulator of SARS-CoV-2-induced male reproductive disruptions. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2022; 27:14. [PMID: 35730047 PMCID: PMC9197722 DOI: 10.1186/s43043-022-00104-8] [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: 01/22/2022] [Accepted: 05/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), causing the coronavirus disease 2019 (COVID-19), has shown its persistent pandemic strength. This viral infectivity, kinetics, and the mechanisms of its actions in human body are still not completely understood. In addition, the infectivity and COVID-19 severity reportedly differ with patient’s gender with men being more susceptible to the disease. Thus, different studies have also suggested the adverse impact of COVID-19 on male reproductive functions, mainly emphasizing on high expressions of angiotensin-converting enzyme 2 (ACE2) in the testes that allows the viral entry into the cells. Main body The N-acetylcysteine (NAC), a potent therapeutic agent of COVID-19, may be effective in reducing the impairing impacts of this disease on male reproductive functions. NAC acts as mucolytic agent by reducing sulfide bonds in the cross-linked glycoprotein matrix in mucus owing to its free sulfhydryl group. Since NAC also breaks the viral disulfide bonds required for the host cell invasion, it may help to prevent direct SARS-CoV-2 invasion into the testicular cells as well. NAC also acts as a potent anti-inflammatory and antioxidant, directly scavenging reactive oxygen species (ROS) and regulating the redox state by maintaining the thiol pool being a precursor of cysteine (an essential substrate for glutathione synthesis). Since it is suggested that male reproductive impairment in COVID-19 patient may be caused by secondary immune responses owing to systemic inflammation and OS, the anti-inflammatory and antioxidant properties of NAC explained above may attribute in protecting the male reproduction functions from these COVID-19-mediated damages. Conclusion This article explains the mechanisms how NAC treatment for COVID-19 may prevent the infection-mediated disruptions in male reproduction.
Collapse
Affiliation(s)
- Pallav Sengupta
- Department of Physiology, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom, Selangor Malaysia
| | - Sulagna Dutta
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Jenjarom, Selangor Malaysia
| |
Collapse
|
26
|
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] [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
|
27
|
COVID-19 and One-Carbon Metabolism. Int J Mol Sci 2022; 23:ijms23084181. [PMID: 35456998 PMCID: PMC9026976 DOI: 10.3390/ijms23084181] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/31/2022] Open
Abstract
Dysregulation of one-carbon metabolism affects a wide range of biological processes and is associated with a number of diseases, including cardiovascular disease, dementia, neural tube defects, and cancer. Accumulating evidence suggests that one-carbon metabolism plays an important role in COVID-19. The symptoms of long COVID-19 are similar to those presented by subjects suffering from vitamin B12 deficiency (pernicious anemia). The metabolism of a cell infected by the SARS-CoV-2 virus is reshaped to fulfill the need for massive viral RNA synthesis, which requires de novo purine biosynthesis involving folate and one-carbon metabolism. Many aspects of host sulfur amino acid metabolism, particularly glutathione metabolism underlying antioxidant defenses, are also taken over by the SARS-CoV-2 virus. The purpose of this review is to summarize recent findings related to one-carbon metabolism and sulfur metabolites in COVID-19 and discuss how they inform strategies to combat the disease.
Collapse
|
28
|
Su H, Liu N, Zhang Y, Kong J. Vitamin D/VDR regulates peripheral energy homeostasis via central renin-angiotensin system. J Adv Res 2021; 33:69-80. [PMID: 34603779 PMCID: PMC8463910 DOI: 10.1016/j.jare.2021.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 01/19/2023] Open
Abstract
Introduction Some epidemiological studies have revealed that vitamin D (VD) deficiency is closely linked with the prevalence of obesity, however, the role of VD in energy homeostasis is yet to be investigated, especially in central nervous system. Given that VD negatively regulates renin in adipose tissue, we hypothesized that central VD might play a potential role in energy homeostasis. Objectives The present study aims to investigate the potential role of VD in energy homeostasis in the CNS and elaborate its underlying mechanisms. Methods This study was conducted in Cyp27b1−/− mice, VD-treated and wild-type mice. After the intraventricular injection of renin or its inhibitors, the changes of renin-angiotensin system (RAS) and its down-stream pathway as well as their effects on metabolic rate were examined. Results The RAS activity was enhanced in Cyp27b1−/− mice, exhibiting a increased metabolic rate. Additionally, corticotropin-releasing hormone (CRH), a RAS-mediated protein regulating energy metabolism in the hypothalamus, increased significantly in Cyp27b1−/− mice. While in VD-treated group, the RAS and sympathetic nerve activities were slightly inhibited, hence the reduced metabolic rate. Conclusion Collectively, the present study demonstrates that the VD/vitamin D receptor (VDR) has a significant impact on energy homeostasis through the modulation of RAS activity in the hypothalamus, subsequently altering CRH expression and sympathetic nervous activity.
Collapse
Affiliation(s)
- Han Su
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ning Liu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yalin Zhang
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Kong
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
29
|
de Miranda FS, Guimarães JPT, Menikdiwela KR, Mabry B, Dhakal R, Rahman RL, Moussa H, Moustaid-Moussa N. Breast cancer and the renin-angiotensin system (RAS): Therapeutic approaches and related metabolic diseases. Mol Cell Endocrinol 2021; 528:111245. [PMID: 33753205 DOI: 10.1016/j.mce.2021.111245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022]
Abstract
The Renin-Angiotensin System (RAS) is classically recognized for regulating blood pressure and fluid balance. Recently, this role has extended to other areas including inflammation, obesity, diabetes, as well as breast cancer. RAS components are expressed in normal and cancerous breast tissues, and downregulation of RAS inhibits metastasis, proliferation, angiogenesis, and desmoplasia in the tumor microenvironment. Therefore, RAS inhibitors (Angiotensin receptor blockers, ARBs, or angiotensin converting enzyme inhibitors, ACE-I) may be beneficial as preventive adjuvant therapies to thwart breast cancer development and improve outcomes, respectively. Given the beneficial effects of RAS inhibitors in metabolic diseases, which often co-exist in breast cancer patients, combining RAS inhibitors with other breast cancer therapies may enhance the effectiveness of current treatments. This review scrutinizes above associations, to advance our understanding of the role of RAS in breast cancer and its potential for repurposing of RAS inhibitors to improve the therapeutic approach for breast cancer patients.
Collapse
Affiliation(s)
- Flávia Sardela de Miranda
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - João Pedro Tôrres Guimarães
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA; Laboratory of Immunopharmacology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo (ICB/USP), São Paulo, SP, Brazil; Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo (FCF/USP), São Paulo, SP, Brazil
| | - Kalhara R Menikdiwela
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Brennan Mabry
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA
| | - Rabin Dhakal
- Department of Mechanical Engineering, Texas Tech University (TTU), Lubbock, TX, USA
| | - Rakhshanda Layeequr Rahman
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Hanna Moussa
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA; Department of Mechanical Engineering, Texas Tech University (TTU), Lubbock, TX, USA
| | - Naima Moustaid-Moussa
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA.
| |
Collapse
|
30
|
Abubakar MB, Usman D, El-Saber Batiha G, Cruz-Martins N, Malami I, Ibrahim KG, Abubakar B, Bello MB, Muhammad A, Gan SH, Dabai AI, Alblihed M, Ghosh A, Badr RH, Thangadurai D, Imam MU. Natural Products Modulating Angiotensin Converting Enzyme 2 (ACE2) as Potential COVID-19 Therapies. Front Pharmacol 2021; 12:629935. [PMID: 34012391 PMCID: PMC8126690 DOI: 10.3389/fphar.2021.629935] [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: 11/16/2020] [Accepted: 03/08/2021] [Indexed: 01/08/2023] Open
Abstract
The 2019 coronavirus disease (COVID-19) is a potentially fatal multisystemic infection caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Currently, viable therapeutic options that are cost effective, safe and readily available are desired, but lacking. Nevertheless, the pandemic is noticeably of lesser burden in African and Asian regions, where the use of traditional herbs predominates, with such relationship warranting a closer look at ethnomedicine. From a molecular viewpoint, the interaction of SARS-CoV-2 with angiotensin converting enzyme 2 (ACE2) is the crucial first phase of COVID-19 pathogenesis. Here, we review plants with medicinal properties which may be implicated in mitigation of viral invasion either via direct or indirect modulation of ACE2 activity to ameliorate COVID-19. Selected ethnomedicinal plants containing bioactive compounds which may prevent and mitigate the fusion and entry of the SARS-CoV-2 by modulating ACE2-associated up and downstream events are highlighted. Through further experimentation, these plants could be supported for ethnobotanical use and the phytomedicinal ligands could be potentially developed into single or combined preventive therapeutics for COVID-19. This will benefit researchers actively looking for solutions from plant bioresources and help lessen the burden of COVID-19 across the globe.
Collapse
Affiliation(s)
- Murtala Bello Abubakar
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Dawoud Usman
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Porto, Portugal
| | - Ibrahim Malami
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
- Department of Pharmacognosy and Ethnopharmacy, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Kasimu Ghandi Ibrahim
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Bilyaminu Abubakar
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Muhammad Bashir Bello
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna Sate, Nigeria
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Aliyu Ibrahim Dabai
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
- Department of Microbiology, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - M Alblihed
- Department of Microbiology, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Arabinda Ghosh
- Microbiology Division, Department of Botany, Gauhati University, Guwahati, India
| | - Reem H. Badr
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | | | - Mustapha Umar Imam
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| |
Collapse
|
31
|
Suganya S, Divya S, Parani M. Severe acute respiratory syndrome-coronavirus-2: Current advances in therapeutic targets and drug development. Rev Med Virol 2021; 31:e2174. [PMID: 32965078 PMCID: PMC7537282 DOI: 10.1002/rmv.2174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/05/2020] [Accepted: 09/08/2020] [Indexed: 12/23/2022]
Abstract
The current pandemic of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) has quickly emerged as a global health concern with government bodies worldwide taking drastic control measures. Understanding the virology of SARS-CoV-2, its molecular mechanisms, and its pathogenesis are required for a targeted therapeutic approach. In this review, we highlight the current molecular and drug advances that target SARS-CoV-2 at the genome level. We also summarize studies that therapeutically target the host angiotensin-converting enzyme 2 and proteases. Finally, we summarize antibody-mediated therapeutic approaches, as well as recent trends in vaccine development. Hence, the purpose of this study is to investigate different molecular targets in SARS-CoV-2 pathogenesis and their usefulness in developing strategies for drug development.
Collapse
Affiliation(s)
- Sakthivel Suganya
- Department of PsychologyTMC AcademySingapore
- Genome Research CenterAcademia SinicaTaipei CityTaiwan, ROC
| | - Suresh Divya
- Department of PediatricsNational Taiwan University HospitalYunlin CountyTaiwan, ROC
- Department of Genetic EngineeringGenomics LaboratorySRM Institute of Science and TechnologyKattankulathurTamil NaduIndia
| | - Madasamy Parani
- Department of Genetic EngineeringGenomics LaboratorySRM Institute of Science and TechnologyKattankulathurTamil NaduIndia
| |
Collapse
|
32
|
Kotru S, Klimuntowski M, Ridha H, Uddin Z, Askhar AA, Singh G, Howlader MMR. Electrochemical sensing: A prognostic tool in the fight against COVID-19. Trends Analyt Chem 2021; 136:116198. [PMID: 33518850 PMCID: PMC7825925 DOI: 10.1016/j.trac.2021.116198] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The COVID-19 pandemic has devastated the world, despite all efforts in infection control and treatment/vaccine development. Hospitals are currently overcrowded, with health statuses of patients often being hard to gauge. Therefore, methods for determining infection severity need to be developed so that high-risk patients can be prioritized, resources can be efficiently distributed, and fatalities can be prevented. Electrochemical prognostic biosensing of various biomarkers may hold promise in solving these problems as they are low-cost and provide timely results. Therefore, we have reviewed the literature and extracted the most promising biomarkers along with their most favourable electrochemical sensors. The biomarkers discussed in this paper are C-reactive protein (CRP), interleukins (ILs), tumour necrosis factor alpha (TNFα), interferons (IFNs), glutamate, breath pH, lymphocytes, platelets, neutrophils and D-dimer. Metabolic syndrome is also discussed as comorbidity for COVID-19 patients, as it increases infection severity and raises chances of becoming infected. Cannabinoids, especially cannabidiol (CBD), are discussed as a potential adjunct therapy for COVID-19 as their medicinal properties may be desirable in minimizing the neurodegenerative or severe inflammatory damage caused by severe COVID-19 infection. Currently, hospitals are struggling to provide adequate care; thus, point-of-care electrochemical sensor development needs to be prioritized to provide an approximate prognosis for hospital patients. During and following the immediate aftermath of the pandemic, electrochemical sensors can also be integrated into wearable and portable devices to help patients monitor recovery while returning to their daily lives. Beyond the COVID-19 pandemic, these sensors will also prove useful for monitoring inflammation-based diseases such as cancer and cardiovascular disease.
Collapse
Affiliation(s)
- Sharda Kotru
- Department of Integrated Biomedical Engineering and Health Sciences, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Martin Klimuntowski
- Department of Electrical and Computer Engineering, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Hashim Ridha
- School of Interdisciplinary Science, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Zakir Uddin
- School of Rehabilitation Science, McMaster University, 1400 Main St W, Hamilton, ON, L8S 1C7, Canada
| | - Ali A Askhar
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| | - Matiar M R Howlader
- Department of Electrical and Computer Engineering, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| |
Collapse
|
33
|
Experimental data using candesartan and captopril indicate no double-edged sword effect in COVID-19. Clin Sci (Lond) 2021; 135:465-481. [PMID: 33479758 PMCID: PMC7851407 DOI: 10.1042/cs20201511] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
The key link between renin–angiotensin system (RAS) and COVID-19 is ACE2 (angiotensin-converting enzyme 2), which acts as a double-edged sword, because ACE2 increases the tissue anti-inflammatory response but it is also the entry receptor for the virus. There is an important controversy on several drugs that regulate RAS activity and possibly ACE2, and are widely used, particularly by patients most vulnerable to severe COVID-19. In the lung of healthy rats, we observed that candesartan (an angiotensin type-1, AT1, receptor blocker; ARB) and captopril (an ACE inhibitor; ACEI) up-regulated expression of tissue ACE2 and RAS anti-inflammatory axis receptors (AT2 and Mas receptors). This effect was particularly pronounced in rats with metabolic syndrome (obesity, increased blood pressure and hyperglycemia) and aged rats. Treatment of cultures of human type-II pneumocytes with candesartan or captopril induced up-regulation of ACE2 expression in cells. Treatment with viral spike protein induced a decrease in full-length (i.e. transmembrane) ACE2, an increase in levels of a short intracellular ACE2 polypeptide and an increase in ADAM17 activity in cells, together with an increase in levels of soluble ACE2 and major proinflammatory cytokines in the culture medium. Spike protein-induced changes and levels of spike protein internalization in cells were inhibited by pretreatment with the above-mentioned drugs. The results suggest that these drugs increase ACE2 levels and promote the anti-inflammatory RAS axis in the lung. Furthermore, possible up-regulation of viral entry by the drug-induced increase in expression of transmembrane ACE2 is counteracted by additional mechanisms, particularly by drug-induced inhibition of ADAM17 activity.
Collapse
|
34
|
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
|
35
|
Moraes DS, Lelis DDF, Andrade JMO, Meyer L, Guimarães ALS, De Paula AMB, Farias LC, Santos SHS. Enalapril improves obesity associated liver injury ameliorating systemic metabolic markers by modulating Angiotensin Converting Enzymes ACE/ACE2 expression in high-fat feed mice. Prostaglandins Other Lipid Mediat 2021; 152:106501. [PMID: 33049402 DOI: 10.1016/j.prostaglandins.2020.106501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 09/24/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Obesity is a chronic disease caused multiple associated factors that results in excessive body fat accumulation. The Renin-Angiotensin System (RAS) unbalance is now recognized as a key factor on regulating body energy and metabolism. AIM The aim of the present study was to evaluate the Enalapril (ACE inhibitor) effects on the metabolic function and hepatic steatosis of obese mice evaluating Angiotensin Converting Enzymes (ACEs) expression. METHODS The experiment was performed using 32 male Swiss mice (8 weeks old) equally and randomly divided into 4 groups (n = 8): standard diet (ST), standard diet plus Enalapril (ST + ENAL), hyperlipidic diet (HF) and hyperlipidic diet plus Enalapril (HF + ENAL). Weekly measurements of animal weight and feed consumption were performed. At the end of treatment period a glucose tolerance test (GTT) and insulin sensitivity test (IST) were performed. Ultrasonography was used to evaluate hepatic and epididymal fat pad. Liver samples were submitted to HE histology and gene expression analyses were performed using Real-Time PCR. RESULTS The main results showed a decrease in body weight after treatment with Enalapril, as well as a reduced size of epididymal fat pad (EFP). Hepatic echogenicity and steatosis measurement were lower in the obese groups treated with Enalapril also modulating ACE2/ACE expressions. CONCLUSIONS Enalapril use improved metabolism reducing hepatic steatosis, decreasing ACE expression and increasing ACE2 expression.
Collapse
Affiliation(s)
- Daniel Silva Moraes
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Deborah de Farias Lelis
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - João Marcus Oliveira Andrade
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Lara Meyer
- Department of Biomedical Sciences, McMurry University, Abilene, TX, USA
| | - André Luiz Sena Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Alfredo Maurício Batista De Paula
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Lucyana Conceição Farias
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa 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
|
36
|
Hypothalamic Renin-Angiotensin System and Lipid Metabolism: Effects of Virgin Olive Oil versus Butter in the Diet. Nutrients 2021; 13:nu13020480. [PMID: 33572630 PMCID: PMC7912484 DOI: 10.3390/nu13020480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/12/2022] Open
Abstract
The brain renin-angiotensin system (RAS) has been recently involved in the homeostatic regulation of energy. Our goal was to analyse the influence of a diet rich in saturated fatty acids (butter) against one enriched in monounsaturated fatty acids (olive oil) on hypothalamic RAS, and their relationship with the metabolism of fatty acids. Increases in body weight and visceral fat, together with an increase in aminopeptidase A expression and reductions in AngII and AngIV were observed in the hypothalamus of animals fed with the butter diet. In this group, a marked reduction in the expression of genes related to lipid metabolism (LPL, CD36, and CPT-1) was observed in liver and muscle. No changes were found in terms of body weight, total visceral fat and the expression of hepatic genes related to fatty acid metabolism in the olive oil diet. The expressions of LPL and CD36 were reduced in the muscles, although the decrease was lower than in the butter diet. At the same time, the fasting levels of leptin were reduced, no changes were observed in the hypothalamic expression of aminopeptidase A and decreases were noted in the levels of AngII, AngIV and AngIII. These results support that the type of dietary fat is able to modify the hypothalamic profile of RAS and the body energy balance, related to changes in lipid metabolism.
Collapse
|
37
|
Nagai N, Kawashima H, Toda E, Homma K, Osada H, Guzman NA, Shibata S, Uchiyama Y, Okano H, Tsubota K, Ozawa Y. Renin-angiotensin system impairs macrophage lipid metabolism to promote age-related macular degeneration in mouse models. Commun Biol 2020; 3:767. [PMID: 33299105 PMCID: PMC7725839 DOI: 10.1038/s42003-020-01483-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 11/16/2020] [Indexed: 12/16/2022] Open
Abstract
Metabolic syndrome, a condition involving obesity and hypertension, increases the risk of aging-associated diseases such as age-related macular degeneration (AMD). Here, we demonstrated that high-fat diet (HFD)-fed mice accumulated oxidized low-density lipoprotein (ox-LDL) in macrophages through the renin–angiotensin system (RAS). The ox-LDL-loaded macrophages were responsible for visual impairment in HFD mice along with a disorder of the retinal pigment epithelium (RPE), which is required for photoreceptor outer segment renewal. RAS repressed ELAVL1, which reduced PPARγ, impeding ABCA1 induction to levels that are sufficient to excrete overloaded cholesterol within the macrophages. The ox-LDL-loaded macrophages expressed inflammatory cytokines and attacked the RPE. An antihypertensive drug, angiotensin II type 1 receptor (AT1R) blocker, resolved the decompensation of lipid metabolism in the macrophages and reversed the RPE condition and visual function in HFD mice. AT1R signaling could be a future therapeutic target for macrophage-associated aging diseases, such as AMD. Nagai et al. show that mice fed high-fat diet (HFD) accumulate oxidized low-density lipoprotein in macrophages through the renin–angiotensin system, which impairs visual function. They find that angiotensin II type 1 receptor (AT1R) improves the visual function of HFD mice, suggesting AT1R signaling as a potential therapeutic target for age-related macular degeneration.
Collapse
Affiliation(s)
- Norihiro Nagai
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan
| | - Hirohiko Kawashima
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan
| | - Eriko Toda
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan
| | - Kohei Homma
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan
| | - Hideto Osada
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan
| | - Naymel A Guzman
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan
| | - Shinsuke Shibata
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan
| | - Yoko Ozawa
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan. .,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan. .,Department of Ophthalmology, St. Luke's International Hospital, 9-1 Akashi-Cho, Chuo-Ku, Tokyo, 104-8560, Japan. .,St. Luke's International University, 9-1 Akashi-Cho, Tokyo, 104-8560, Japan.
| |
Collapse
|
38
|
Lim I, Mitsui R, Kameda M, Sellers DJ, Chess-Williams R, Hashitani H. Comparative effects of angiotensin II on the contractility of muscularis mucosae and detrusor in the pig urinary bladder. Neurourol Urodyn 2020; 40:102-111. [PMID: 33074588 DOI: 10.1002/nau.24548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 01/20/2023]
Abstract
To explore contractile actions of angiotensin II (ATII) on the muscularis mucosae (MM) of the bladder, ATII-induced contractions were compared between MM and the detrusor smooth muscle (DSM) of the pig bladder by isometric tension recordings. Effects of ATII on spontaneous Ca2+ transients in MM were visualized using Cal-520 fluorescence. ATII receptor type 1 (ATR1) expression in MM and DSM was also examined by immunohistochemistry. ATII (1 nM-1 μM) caused phasic contractions of MM in a concentration-dependent manner, while ATII (10 nM-10 μM) had no or marginal effects on DSM contractility. ATII (100 nM)-induced MM contractions had an amplitude of approximately 70% of carbachol (1 μM)-induced or 90% of U46619 (100 nM)-induced contractions. Candesartan (10 nM), an ATR1 blocker, prevented the contractile effects of ATII (1 nM) in MM, while ATR1 immunofluorescence was greater in MM than DSM. ATII (10-100 pM) increased the frequency but not the amplitude of spontaneous Ca2+ transients in MM. Both urothelium-intact and -denuded MM strips developed comparable spontaneous phasic contractions, but ATII, carbachol and U46619-induced contractions were significantly larger in urothelium-denuded than urothelium-intact MM strips. In conclusion, the MM appears to have a much greater sensitivity to ATII compared with DSM that could well sense circulating ATII, suggesting that MM may be the predominant target of contractile actions induced by ATII in the bladder while the urothelium appears to inhibit MM contractility.
Collapse
Affiliation(s)
- Iris Lim
- Center for Urology, Faculty of Health Sciences & Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Retsu Mitsui
- Department of Cell Physiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masashi Kameda
- Department of Cell Physiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Donna Jayne Sellers
- Center for Urology, Faculty of Health Sciences & Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Russ Chess-Williams
- Center for Urology, Faculty of Health Sciences & Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Hikaru Hashitani
- Department of Cell Physiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
39
|
Giori IG, Magliano DC, Alexandre-Santos B, Fernandes T, Oliveira EM, Vieira CP, Conte-Junior CA, Ceddia RB, Nobrega ACL, Frantz EDC. Enalapril and treadmill running reduce adiposity, but only the latter causes adipose tissue browning in mice. J Cell Physiol 2020; 236:900-910. [PMID: 32617979 DOI: 10.1002/jcp.29900] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/18/2022]
Abstract
This study investigated whether regulation of the renin-angiotensin system (RAS) by enalapril and/or aerobic exercise training (AET) causes browning of the subcutaneous white adipose tissue (sWAT). C57BL/6 mice were fed either a standard chow or a high-fat (HF) diet for 16 weeks. At Week 8, HF-fed animals were divided into sedentary (HF), enalapril (HF-E), AET (HF-T), and enalapril plus AET (HF-ET) groups. Subsequently, sWAT was extracted for morphometry, determination of RAS expression, and biomarkers of WAT browning. The HF group displayed adipocyte hypertrophy and induction of the classical RAS axis. Conversely, all interventions reduced adiposity and induced the counterregulatory RAS axis. However, only AET raised plasma irisin, increased peroxisome proliferator-activated receptor-γ coactivator-1α, and uncoupling protein-1 levels, and the expression of PR-domain containing 16 in sWAT. Therefore, we concluded that AET-induced sWAT browning was independent of the counterregulatory axis shifting of RAS in HF diet-induced obesity.
Collapse
Affiliation(s)
- Isabele G Giori
- Laboratory of Exercise Sciences, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - D'Angelo C Magliano
- Laboratory of Morphological and Metabolic Analyses, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Beatriz Alexandre-Santos
- Laboratory of Exercise Sciences, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil.,Laboratory of Morphological and Metabolic Analyses, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Tiago Fernandes
- National Institute for Science and Technology, INCT Physical (In)activity and Exercise, CNPq, Niteroi, Rio de Janeiro, Brazil.,Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, São Paulo, State of São Paulo, Brazil
| | - Edilamar M Oliveira
- National Institute for Science and Technology, INCT Physical (In)activity and Exercise, CNPq, Niteroi, Rio de Janeiro, Brazil.,Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, São Paulo, State of São Paulo, Brazil
| | - Carla P Vieira
- Department of Food Technology, Faculty of Veterinary, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Carlos A Conte-Junior
- Department of Food Technology, Faculty of Veterinary, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Rolando B Ceddia
- Muscle Health Research Center, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Antonio C L Nobrega
- Laboratory of Exercise Sciences, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil.,National Institute for Science and Technology, INCT Physical (In)activity and Exercise, CNPq, Niteroi, Rio de Janeiro, Brazil
| | - Eliete D C Frantz
- Laboratory of Exercise Sciences, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil.,Laboratory of Morphological and Metabolic Analyses, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil.,National Institute for Science and Technology, INCT Physical (In)activity and Exercise, CNPq, Niteroi, Rio de Janeiro, Brazil
| |
Collapse
|
40
|
Samavati L, Uhal BD. ACE2, Much More Than Just a Receptor for SARS-COV-2. Front Cell Infect Microbiol 2020; 10:317. [PMID: 32582574 PMCID: PMC7294848 DOI: 10.3389/fcimb.2020.00317] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/26/2020] [Indexed: 12/17/2022] Open
Abstract
The rapidly evolving pandemic of severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection worldwide cost many lives. The angiotensin converting enzyme-2 (ACE-2) has been identified as the receptor for the SARS-CoV-2 viral entry. As such, it is now receiving renewed attention as a potential target for anti-viral therapeutics. We review the physiological functions of ACE2 in the cardiovascular system and the lungs, and how the activation of ACE2/MAS/G protein coupled receptor contributes in reducing acute injury and inhibiting fibrogenesis of the lungs and protecting the cardiovascular system. In this perspective, we predominantly focus on the impact of SARS-CoV-2 infection on ACE2 and dysregulation of the protective effect of ACE2/MAS/G protein pathway vs. the deleterious effect of Renin/Angiotensin/Aldosterone. We discuss the potential effect of invasion of SARS-CoV-2 on the function of ACE2 and the loss of the protective effect of the ACE2/MAS pathway in alveolar epithelial cells and how this may amplify systemic deleterious effect of renin-angiotensin aldosterone system (RAS) in the host. Furthermore, we speculate the potential of exploiting the modulation of ACE2/MAS pathway as a natural protection of lung injury by modulation of ACE2/MAS axis or by developing targeted drugs to inhibit proteases required for viral entry.
Collapse
Affiliation(s)
- Lobelia Samavati
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine and Detroit Medical Center, Wayne State University, Detroit, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Bruce D. Uhal
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
41
|
Menikdiwela KR, Ramalingam L, Rasha F, Wang S, Dufour JM, Kalupahana NS, Sunahara KKS, Martins JO, Moustaid-Moussa N. Autophagy in metabolic syndrome: breaking the wheel by targeting the renin-angiotensin system. Cell Death Dis 2020; 11:87. [PMID: 32015340 PMCID: PMC6997396 DOI: 10.1038/s41419-020-2275-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/12/2022]
Abstract
Metabolic syndrome (MetS) is a complex, emerging epidemic which disrupts the metabolic homeostasis of several organs, including liver, heart, pancreas, and adipose tissue. While studies have been conducted in these research areas, the pathogenesis and mechanisms of MetS remain debatable. Lines of evidence show that physiological systems, such as the renin-angiotensin system (RAS) and autophagy play vital regulatory roles in MetS. RAS is a pivotal system known for controlling blood pressure and fluid balance, whereas autophagy is involved in the degradation and recycling of cellular components, including proteins. Although RAS is activated in MetS, the interrelationship between RAS and autophagy varies in glucose homeostatic organs and their cross talk is poorly understood. Interestingly, autophagy is attenuated in the liver during MetS, whereas autophagic activity is induced in adipose tissue during MetS, indicating tissue-specific discordant roles. We discuss in vivo and in vitro studies conducted in metabolic tissues and dissect their tissue-specific effects. Moreover, our review will focus on the molecular mechanisms by which autophagy orchestrates MetS and the ways future treatments could target RAS in order to achieve metabolic homeostasis.
Collapse
Affiliation(s)
- Kalhara R Menikdiwela
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Fahmida Rasha
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Shu Wang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Jannette M Dufour
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Nishan S Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
- Department of Physiology, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Karen K S Sunahara
- Department of Experimental Physiopatholgy, Medical School University of São Paulo, São Paulo, Brazil
| | - Joilson O Martins
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences of University Sao Paulo (FCF/USP), São Paulo, Brazil
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA.
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA.
| |
Collapse
|
42
|
Vargas-Castillo A, Tobon-Cornejo S, Del Valle-Mondragon L, Torre-Villalvazo I, Schcolnik-Cabrera A, Guevara-Cruz M, Pichardo-Ontiveros E, Fuentes-Romero R, Bader M, Alenina N, Vidal-Puig A, Hong E, Torres N, Tovar AR. Angiotensin-(1-7) induces beige fat thermogenesis through the Mas receptor. Metabolism 2020; 103:154048. [PMID: 31843339 DOI: 10.1016/j.metabol.2019.154048] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Angiotensin-(1-7) [Ang-(1-7)], a component of the renin angiotensin system, is a vasodilator that exerts its effects primarily through the Mas receptor. The discovery of the Mas receptor in white adipose tissue (WAT) suggests an additional role for this peptide. The aim of the present study was to assess whether Ang-(1-7) can induce the expression of thermogenic genes in white adipose tissue and increase mitochondrial respiration in adipocytes. MATERIALS/METHODS Stromal Vascular fraction (SVF)-derived from mice adipose tissue was stimulated for one week with Ang-(1-7), then expression of beige markers and mitochondrial respiration were assessed. Mas+/+ and Mas-/- mice fed a control diet or a high fat-sucrose diet (HFSD) were exposed to a short or long term infusion of Ang-(1-7) and body weight, body fat, energy expenditure, cold resistance and expression of beige markers were assessed. Also, transgenic rats overexpressing Ang-(1-7) were fed with a control diet or a high fat-sucrose diet and the same parameters were assessed. Ang-(1-7) circulating levels from human subjects with different body mass index (BMI) or age were measured. RESULTS Incubation of adipocytes derived from SVF with Ang-(1-7) increased the expression of beige markers. Infusion of Ang-(1-7) into lean and obese Mas+/+mice also induced the expression of Ucp1 and some beige markers, an effect not observed in Mas-/- mice. Mas-/- mice had increased body weight gain and decreased cold resistance, whereas rats overexpressing Ang-(1-7) showed the opposite effects. Overexpressing rats exposed to cold developed new thermogenic WAT in the anterior interscapular area. Finally, in human subjects the higher the BMI, low circulating concentration of Ang-(1-7) levels were detected. Similarly, the circulating levels of Ang-(1-7) peptide were reduced with age. CONCLUSION These data indicate that Ang-(1-7) stimulates beige markers and thermogenesis via the Mas receptor, and this evidence suggests a potential therapeutic use to induce thermogenesis of WAT, particularly in obese subjects that have reduced circulating concentration of Ang-(1-7).
Collapse
MESH Headings
- Adipose Tissue, Beige/drug effects
- Adipose Tissue, Beige/metabolism
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adult
- Angiotensin I/pharmacology
- Animals
- Cell Respiration/drug effects
- Cell Respiration/genetics
- Cells, Cultured
- Energy Metabolism/genetics
- Female
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Middle Aged
- Obesity/genetics
- Obesity/metabolism
- Obesity/pathology
- Peptide Fragments/pharmacology
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/physiology
- Rats
- Rats, Transgenic
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/physiology
- Thermogenesis/drug effects
- Thermogenesis/genetics
- Young Adult
Collapse
Affiliation(s)
- Ariana Vargas-Castillo
- Departmento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; Deparmento de Farmacobiología, Centro de Investigación y de Estudios Avanzados Sede Sur, Ciudad de México 14330, Mexico
| | - Sandra Tobon-Cornejo
- Departmento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | | | - Ivan Torre-Villalvazo
- Departmento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Alejandro Schcolnik-Cabrera
- Departmento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Martha Guevara-Cruz
- Departmento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Edgar Pichardo-Ontiveros
- Departmento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Rebeca Fuentes-Romero
- Departmento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine, Berlin 13092, Germany; Charité University Medicine Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany; Institute for Biology, University of Lübeck, Germany
| | - Natalia Alenina
- Max Delbrück Center for Molecular Medicine, Berlin 13092, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom
| | - Enrique Hong
- Deparmento de Farmacobiología, Centro de Investigación y de Estudios Avanzados Sede Sur, Ciudad de México 14330, Mexico
| | - Nimbe Torres
- Departmento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Armando R Tovar
- Departmento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico.
| |
Collapse
|
43
|
Samavati L, Uhal BD. ACE2, Much More Than Just a Receptor for SARS-COV-2. Front Cell Infect Microbiol 2020. [PMID: 32582574 DOI: 10.3389/fcimb.2020.0317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
The rapidly evolving pandemic of severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection worldwide cost many lives. The angiotensin converting enzyme-2 (ACE-2) has been identified as the receptor for the SARS-CoV-2 viral entry. As such, it is now receiving renewed attention as a potential target for anti-viral therapeutics. We review the physiological functions of ACE2 in the cardiovascular system and the lungs, and how the activation of ACE2/MAS/G protein coupled receptor contributes in reducing acute injury and inhibiting fibrogenesis of the lungs and protecting the cardiovascular system. In this perspective, we predominantly focus on the impact of SARS-CoV-2 infection on ACE2 and dysregulation of the protective effect of ACE2/MAS/G protein pathway vs. the deleterious effect of Renin/Angiotensin/Aldosterone. We discuss the potential effect of invasion of SARS-CoV-2 on the function of ACE2 and the loss of the protective effect of the ACE2/MAS pathway in alveolar epithelial cells and how this may amplify systemic deleterious effect of renin-angiotensin aldosterone system (RAS) in the host. Furthermore, we speculate the potential of exploiting the modulation of ACE2/MAS pathway as a natural protection of lung injury by modulation of ACE2/MAS axis or by developing targeted drugs to inhibit proteases required for viral entry.
Collapse
Affiliation(s)
- Lobelia Samavati
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine and Detroit Medical Center, Wayne State University, Detroit, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Bruce D Uhal
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
44
|
Adipocytes initiate an adipose-cerebral-peripheral sympathetic reflex to induce insulin resistance during high-fat feeding. Clin Sci (Lond) 2019; 133:1883-1899. [PMID: 31477624 DOI: 10.1042/cs20190412] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/06/2019] [Accepted: 09/02/2019] [Indexed: 01/22/2023]
Abstract
The underlying mechanism by which amassing of white adipose tissue in obesity regulates sympathetic nerve system (SNS) drive to the tissues responsible for glucose disposal, and causes insulin resistance (IR), remains unknown. We tested the hypothesis that high-fat (HF) feeding increases afferent impulses from white adipose tissue that reflexively elevate efferent nerve activity to skeletal muscle (SM) and adipose tissue to impair their local glucose uptake. We also investigated how salt-intake can enhance IR. HF-fed rats received a normal salt (0.4%) or high salt (4%) diet for 3 weeks. High-salt intake in HF fed rats decreased insulin-stimulated 2-deoxyglucose uptake by over 30% in white adipose tissue and SM, exacerbated inflammation, and impaired their insulin signaling and glucose transporter 4 (Glut4) trafficking. Dietary salt in HF fed rats also increased the activity of the adipose-cerebral-muscle renin-angiotensin system (RAS) axes, SNS, and reactive oxygen species (ROS). Insulin sensitivity was reduced by 32% in HF rats during high-salt intake, but was improved by over 62% by interruption of central RAS and SNS drive, and by over 45% by denervation or deafferentation of epididymal fat (all P<0.05). Our study suggest that a HF diet engages a sympathetic reflex from the white adipose tissue that activates adipose-cerebral-muscle RAS/ROS axes and coordinates a reduction in peripheral glucose uptake. These are all enhanced by salt-loading. These findings provide new insight into the role of a reflex initiated in adipose tissue in the regulation of glucose homeostasis during HF feeding that could lead to new therapeutic approaches to IR.
Collapse
|
45
|
Glover Z, Hodges MD, Dravecz N, Cameron J, Askwith H, Shirras A, Broughton SJ. Loss of angiotensin-converting enzyme-related (ACER) peptidase disrupts behavioural and metabolic responses to diet in Drosophila melanogaster. ACTA ACUST UNITED AC 2019; 222:jeb.194332. [PMID: 30940674 DOI: 10.1242/jeb.194332] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 03/29/2019] [Indexed: 12/15/2022]
Abstract
Drosophila Acer (Angiotensin-converting enzyme-related) encodes a member of the angiotensin-converting enzyme (ACE) family of metallopeptidases that in mammals play roles in the endocrine regulation of blood homeostasis. ACE is also expressed in adipose tissue, where it is thought to play a role in metabolic regulation. Drosophila ACER is expressed in the adult fat body of the head and abdomen and is secreted into the haemolymph. Acer null mutants have previously been found to have reduced night-time sleep and greater sleep fragmentation. ACER may thus be part of a signalling system linking metabolism with sleep. To further understand the role of ACER in response to diet, we measured sleep and other nutrient-responsive phenotypes in Acer null flies under different dietary conditions. We show that loss of Acer disrupts the normal response of sleep to changes in nutrition. Other nutrient-sensitive phenotypes, including survival and glycogen storage, were also altered in the Acer mutant but lipid storage was not. Although the physiological substrate of the ACER peptidase has not been identified, an alteration of the normal nutrient-dependent control of Drosophila insulin-like peptide 5 protein in the Acer mutant suggests insulin/IGF-like signalling as a candidate pathway modulated by ACER in the nutrient-dependent control of sleep, survival and metabolism.
Collapse
Affiliation(s)
- Zoe Glover
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Matthew D Hodges
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Nikolett Dravecz
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Jack Cameron
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Helen Askwith
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Alan Shirras
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Susan J Broughton
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| |
Collapse
|
46
|
El-Kabbany ZA, Hamza RT, Shinkar DM, Kamal TM, Abdelmageed RI, Said MS, Abdel-Hamid MI. Screening of Egyptian obese children and adolescents for insertion/deletion (I/D) polymorphism in angiotensin-converting enzyme gene. Int J Pediatr Adolesc Med 2019; 6:21-24. [PMID: 31304224 PMCID: PMC6603068 DOI: 10.1016/j.ijpam.2019.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/31/2019] [Accepted: 02/21/2019] [Indexed: 11/16/2022]
Abstract
Background /aims: The role of angiotensin-converting enzyme (ACE) gene polymorphism in the development of obesity and hypertension in children has not been widely studied. We aimed to screen Egyptian obese children and adolescents for insertion/deletion (I/D) polymorphism in the ACE gene. Methods One hundred forty-two children and adolescents were included (70 with simple obesity and 72 controls). Blood pressure was measured, and anthropometric parameters were assessed in all included children and adolescents. Fasting lipid profile, fasting glucose, and insulin were measured. DNA extraction and ACE I/D polymorphism genotyping were also performed. Results Obese children had a higher frequency of DD genotype (30% in obese versus 11.1% in controls, P = .01) and D alleles (61.8% in obese versus 48.6% in controls, P = .01). Obese children with hypertension and prehypertension had higher frequency of DD genotype than II genotype and higher D alleles than I alleles. DD genotype and D allele were independently associated with hypertension (OR: 9.86 and 11.57, respectively, P < .001), while dyslipidemia and insulin resistance were not associated with the ACE I/D gene polymorphism. Conclusion DD genotype and D-allele of the ACE gene polymorphism were associated with obesity and with hypertension and pre-hypertension in Egyptian children.
Collapse
Affiliation(s)
| | - Rasha T. Hamza
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Dina M. Shinkar
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Corresponding author: Dina M Shinkar. Department of Pediatrics, Faculty of Medicine, Ain Shams University, Abbassya Square, Cairo, Egypt. Tel.: +201112636097.
| | - Tarek M. Kamal
- Human Medical Genetics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | | |
Collapse
|
47
|
Barbosa MA, de Sousa GG, de Castro UGM, Carneiro CM, Figueiredo VP, Guerra de Sá R, Santos RASD, Alzamora AC. Oral Ang-(1-7) treatment improves white adipose tissue remodeling and hypertension in rats with metabolic syndrome. Nutrition 2019; 67-68S:100004. [PMID: 34332714 DOI: 10.1016/j.nutx.2019.100004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/01/2019] [Accepted: 10/05/2019] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Angiotensin (Ang)-(1-7) has preventive effects on metabolic syndrome (MetS). The aim of this study was to evaluate the therapeutic effect of oral Ang-(1-7) on mean arterial pressure (MAP), insulin resistance (IR), inflammatory process, and remodeling of white adipose tissue (WAT) in rats with established MetS. METHODS Rats were subjected to control (CT; AIN-93M) or high-fat (HF) diets for 13 wk to induce MetS and treated with Ang-(1-7) or vehicle (V) for the last 6 wk. At the end of 13 wk, MAP, biochemical and histological parameters, and uncoupling protein (UCP) and inflammatory gene expression were determined by quantitative reverse transcription polymerase chain reaction. RESULTS HF-V rats showed increased visceral fat deposition, inflammatory cytokine expression, hyperplasia, and hypertrophy in retroperitoneal (WAT) and brown adipose tissue (BAT). Additionally, the gastrocnemius muscle reduced UCP-3 and increased the UCP-1 expression in BAT. HF-V also elevated levels of plasma insulin, glucose, homeostatic model assessment (HOMA) of IR and HOMA-β, and increased body mass, adiposity, and MAP. Ang-(1-7) treatment in rats with MetS [HF-Ang-(1-7)] reduced WAT area, number of adipocytes, and expression of proinflammatory adipokines in WAT and BAT and increased UCP-3 in gastrocnemius muscle and UCP-1 expression in BAT compared with the HF-V group. These events prevented body mass gain, reduced adiposity, and normalized fasting plasma glucose, insulin levels, HOMA-IR, HOMA-β, and MAP. CONCLUSION Data from the present study demonstrated that oral Ang-(1-7) treatment is effective in restoring biochemical parameters and hypertension in established MetS by improving hypertrophy and hyperplasia in WAT and inflammation in adipose tissue, and regulating metabolic processes in the gastrocnemius muscle and BAT.
Collapse
Affiliation(s)
- Maria Andréa Barbosa
- Núcleo de Pesquisa em Ciências Biológicas, Universidade Federal de Ouro Preto, MG, Brazil
| | | | | | | | | | - Renata Guerra de Sá
- Núcleo de Pesquisa em Ciências Biológicas, Universidade Federal de Ouro Preto, MG, Brazil; Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas Ouro Preto, MG, Brazil
| | | | - Andréia Carvalho Alzamora
- Núcleo de Pesquisa em Ciências Biológicas, Universidade Federal de Ouro Preto, MG, Brazil; Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas Ouro Preto, MG, Brazil.
| |
Collapse
|
48
|
Petek B, Villa-Lopez M, Loera-Valencia R, Gerenu G, Winblad B, Kramberger MG, Ismail MAM, Eriksdotter M, Garcia-Ptacek S. Connecting the brain cholesterol and renin-angiotensin systems: potential role of statins and RAS-modifying medications in dementia. J Intern Med 2018; 284:620-642. [PMID: 30264910 DOI: 10.1111/joim.12838] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Millions of people worldwide receive agents targeting the renin-angiotensin system (RAS) to treat hypertension or statins to lower cholesterol. The RAS and cholesterol metabolic pathways in the brain are autonomous from their systemic counterparts and are interrelated through the cholesterol metabolite 27-hydroxycholesterol (27-OHC). These systems contribute to memory and dementia pathogenesis through interference in the amyloid-beta cascade, vascular mechanisms, glucose metabolism, apoptosis, neuroinflammation and oxidative stress. Previous studies examining the relationship between these treatments and cognition and dementia risk have produced inconsistent results. Defining the blood-brain barrier penetration of these medications has been challenging, and the mechanisms of action on cognition are not clearly established. Potential biases are apparent in epidemiological and clinical studies, such as reverse epidemiology, indication bias, problems defining medication exposure, uncertain and changing doses, and inappropriate grouping of outcomes and medications. This review summarizes current knowledge of the brain cholesterol and RAS metabolism and the mechanisms by which these pathways affect neurodegeneration. The putative mechanisms of action of statins and medications inhibiting the RAS will be examined, together with prior clinical and animal studies on their effects on cognition. We review prior epidemiological studies, analysing their strengths and biases, and identify areas for future research. Understanding the pathophysiology of the brain cholesterol system and RAS and their links to neurodegeneration has enormous potential. In future, well-designed epidemiological studies could identify potential treatments for Alzheimer's disease (AD) amongst medications that are already in use for other indications.
Collapse
Affiliation(s)
- B Petek
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, University Medical Centre, Ljubljana, Slovenia.,University of Ljubljana, Ljubljana, Slovenia
| | - M Villa-Lopez
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - R Loera-Valencia
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - G Gerenu
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neurosciences, Biodonostia Health Research Institute, San Sebastian, Spain.,Center for Networked Biomedical Research in Neurodegenerative Diseases, CIBERNED, Health Institute Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - B Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - M G Kramberger
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, University Medical Centre, Ljubljana, Slovenia.,University of Ljubljana, Ljubljana, Slovenia
| | - M-A-M Ismail
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Neuro, Diseases of the Nervous System patient flow, Karolinska University Hospital, Huddinge, Sweden
| | - M Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - S Garcia-Ptacek
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Internal Medicine, Neurology Section, Södersjukhuset, Stockholm, Sweden
| |
Collapse
|
49
|
Xie CL, Kang SS, Lu C, Choi YJ. Quantification of Multifunctional Dipeptide YA from Oyster Hydrolysate for Quality Control and Efficacy Evaluation. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8437379. [PMID: 30345307 PMCID: PMC6174814 DOI: 10.1155/2018/8437379] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/23/2018] [Accepted: 09/04/2018] [Indexed: 02/07/2023]
Abstract
YA is an angiotensin-I-converting enzyme- (ACE-) inhibitory peptide from oyster hydrolysate with antihypertensive activity. Its antioxidant and anti-inflammatory activity were investigated in this study. YA can dose-dependently quench DPPH and ABTS radical and inhibit lipopolysaccharide-induced nitric oxide in RAW 264.7 cells. YA is a multifunctional peptide and was selected as an indicator for quality control and efficacy evaluation of oyster hydrolysate. A practical HPLC/UV assay for YA quantification was developed and validated. It was proved to be accurate and reliable, according to parameters such as specificity, linearity, precision, and accuracy. The quantity results of YA showed that the stage of enzymatic hydrolysis was a critical control point for quality control; the efficacy of oyster hydrolysate can be enhanced after digested in the gastrointestinal tract due to the release of YA by brush border peptidases. Therefore, YA from oyster hydrolysate is a potential bioactive ingredient for functional foods to combat hypertension.
Collapse
Affiliation(s)
- Cheng-Liang Xie
- Department of Medical Statistics and Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangdong Engineering Technology Research Center of Nutrition Translation, School of Public Health, Sun Yat-Sen University, Guangzhou 510000, China
| | - Sang Soo Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Ciyong Lu
- Department of Medical Statistics and Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangdong Engineering Technology Research Center of Nutrition Translation, School of Public Health, Sun Yat-Sen University, Guangzhou 510000, China
| | - Yeung Joon Choi
- Department of Seafood Science and Technology/Institute of Marine Industry, Gyeongsang National University, Tongyeong 53064, Republic of Korea
| |
Collapse
|
50
|
Purification and identification of adipogenic-differentiating peptides from egg white hydrolysate. Food Chem 2018; 259:25-30. [DOI: 10.1016/j.foodchem.2018.03.099] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 03/02/2018] [Accepted: 03/22/2018] [Indexed: 12/11/2022]
|