1
|
Ivatt L, Paul M, Miguelez-Crespo A, Hadoke PWF, Bailey MA, Morgan RA, Nixon M. Obesity-induced mesenteric PVAT remodelling is sexually dimorphic, but not driven by ovarian hormones : Short title: Obesity induces sex-specific responses in mesenteric PVAT. Cardiovasc Diabetol 2025; 24:39. [PMID: 39856754 PMCID: PMC11762466 DOI: 10.1186/s12933-025-02596-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Obesity, a major risk factor for cardiovascular disease (CVD), is associated with hypertension and vascular dysfunction. Perivascular adipose tissue (PVAT), a metabolically active tissue surrounding blood vessels, plays a key role in regulating vascular tone. In obesity, PVAT becomes dysregulated which may contribute to vascular dysfunction; how sex impacts the remodelling of PVAT and thus the altered vascular contractility during obesity is unclear. OBJECTIVE To investigate sex-specific PVAT dysregulation in the setting of obesity as a potential driver of sex differences in vascular pathologies and CVD risk. METHODS Adult male and female C57Bl/6J mice were fed an obesogenic high-fat diet (HFD) or regular chow for 16 weeks. Mesenteric PVAT (mPVAT) was isolated for RNA-sequencing and histological analysis, and mesenteric arteries were isolated for assessment of vascular function by wire myography. In a separate study, female mice were subjected to bilateral ovariectomy prior to dietary intervention to determine the contribution of ovarian hormones to PVAT dysregulation. RESULTS Transcriptomic analysis of mPVAT revealed sexually dimorphic responses to HFD, with upregulation of extracellular matrix (ECM) remodelling pathways in male but not female mice. Histological and RT-qPCR approaches demonstrated increased collagen deposition and ECM remodelling in mPVAT from obese male compared with obese female mice. Assessment of vascular function in mesenteric arteries -/+ PVAT revealed that in obesity, mPVAT impaired endothelium-mediated vasodilation in male but not female mice. Ovariectomy of female mice prior to HFD administration did not alter ECM transcript expression or collagen deposition in mPVAT compared to sham-operated female mice. CONCLUSIONS Obesity induces sex-specific molecular remodelling in mPVAT, with male mice exhibiting unique upregulation of ECM pathways and increased collagen deposition compared to females. Moreover, the relative protection of female mice from obesity-induced mPVAT dysregulation is not mediated by ovarian hormones. These data highlight a potential sex-specific mechanistic link between mPVAT and mesenteric artery dysfunction in obesity, and provides crucial insights for future development of treatment strategies that consider the unique cardiovascular risks in men and women.
Collapse
Affiliation(s)
- Lisa Ivatt
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Mhairi Paul
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | | | - Patrick W F Hadoke
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Matthew A Bailey
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | | | - Mark Nixon
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK.
| |
Collapse
|
2
|
Steiger S, Li L, Bruchfeld A, Stevens KI, Moran SM, Floege J, Caravaca-Fontán F, Mirioglu S, Teng OYK, Frangou E, Kronbichler A. Sex dimorphism in kidney health and disease: mechanistic insights and clinical implication. Kidney Int 2025; 107:51-67. [PMID: 39477067 DOI: 10.1016/j.kint.2024.08.038] [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: 06/10/2024] [Revised: 07/16/2024] [Accepted: 08/09/2024] [Indexed: 11/18/2024]
Abstract
Sex is a key variable in the regulation of human physiology and pathology. Many diseases disproportionately affect one sex: autoimmune diseases, such as systemic lupus erythematosus, are more common in women but more severe in men, whereas the incidence of other disorders such as gouty arthritis and malignant cancers is higher in men. Besides the pathophysiology, sex may also influence the efficacy of therapeutics; participants in clinical trials are still predominately men, and the side effects of drugs are more common in women than in men. Sex dimorphism is a prominent feature of kidney physiology and function, and consequently affects the predisposition to many adult kidney diseases. These differences subsequently influence the response to immune stimuli, hormones, and therapies. It is highly likely that these responses differ between the sexes. Therefore, it becomes imperative to consider sex differences in translational science from basic science to preclinical research to clinical research and trials. Under-representation of one sex in preclinical animal studies or clinical trials remains an issue and key reported outcomes of such studies ought to be presented separately. Without this, it remains difficult to tailor the management of kidney disease appropriately and effectively. In this review, we provide mechanistic insights into sex differences in rodents and humans, both in kidney health and disease, highlight the importance of considering sex differences in the design of any preclinical animal or clinical study, and propose guidance on how to optimal design and conduct preclinical animal studies in future research.
Collapse
Affiliation(s)
- Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital Munich, Munich, Germany.
| | - Li Li
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital Munich, Munich, Germany
| | - Annette Bruchfeld
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden; Department of Renal Medicine, Karolinska University Hospital and CLINTEC Karolinska Institutet, Stockholm, Sweden
| | - Kate I Stevens
- Glasgow Renal and Transplant Unit, Queen Elizabeth University Hospital, Glasgow, UK
| | - Sarah M Moran
- Cork University Hospital, University College Cork, Cork, Ireland
| | - Jürgen Floege
- Division of Nephrology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University Hospital, Aachen, Germany
| | - Fernando Caravaca-Fontán
- Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain; Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Safak Mirioglu
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Instanbul, Turkey
| | - Onno Y K Teng
- Center of Expertise for Lupus, Vasculitis and Complement-mediated Systemic disease (LuVaCs), Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Eleni Frangou
- Department of Nephrology, Limassol General Hospital, Limassol, Cyprus; University of Nicosia Medical School, Nicosia, Cyprus; National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Andreas Kronbichler
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
3
|
Alhashim A, Capehart K, Tang J, Saad KM, Abdelsayed R, Cooley MA, Williams JM, Elmarakby AA. Does Sex Matter in Obesity-Induced Periodontal Inflammation in the SS LepR Mutant Rats? Dent J (Basel) 2024; 13:14. [PMID: 39851590 PMCID: PMC11764266 DOI: 10.3390/dj13010014] [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: 10/09/2024] [Revised: 12/05/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025] Open
Abstract
Introduction: The incidence of obesity has dramatically increased worldwide. Obesity has been shown to exacerbate the progression of periodontal disease. Studies suggest a sex difference in periodontitis, whereby males are more sensitive to periodontal inflammation compared to females. Aim: In the current study, it was hypothesized that obesity drives periodontal inflammation and bone loss in both sexes. Methodology: Utilizing leptin receptor mutant (SSLepR mutant) rats as a genetic model of obesity, 11-12-week-old male and female lean Dahl salt-sensitive (SS) rats and obese SSLepR mutant rats were used to investigate sex differences in obesity-induced periodontal inflammation. Results: Body weight, insulin, hemoglobin A1c and cholesterol levels were significantly elevated in the obese SSLepR mutant strain vs. the lean SS strain within the same sex. Sex differences in body weight and plasma hemoglobin A1c were only observed in obese SSLepR mutant rats, with males having significantly greater body weight and hemoglobin A1c vs. females. Plasma thiobarbituric acid reactive substances (TBARs) and monocyte chemoattractant protein-1 (MCP-1), markers of systemic oxidative stress and inflammation, respectively, were significantly elevated in obese SSLepR mutant rats vs. lean SS rats, with no sex differences in these parameters in either rat strains. Although micro-CT analyses of the maxillary first molar alveolar bone from obese SSLepR mutant rats revealed no evidence of bone loss and/or sex differences, immuno-histochemical analysis revealed significant elevations in periodontal IL-6 and decreases in IL-10 in obese SSLepR mutant rats vs. lean SS rats, with no apparent sex differences in these parameters. Conclusions: Obesity increases systemic and periodontal inflammation, without evidence of bone loss or apparent sex differences in SSLepR mutant rats.
Collapse
Affiliation(s)
- Abdulmohsin Alhashim
- Departments of General Dentistry and Oral Biology & Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.A.); (K.C.)
| | - Kim Capehart
- Departments of General Dentistry and Oral Biology & Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.A.); (K.C.)
| | - Jocelyn Tang
- Oral Biology & Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.T.); (K.M.S.); (R.A.); (M.A.C.)
| | - Karim M. Saad
- Oral Biology & Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.T.); (K.M.S.); (R.A.); (M.A.C.)
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Rafik Abdelsayed
- Oral Biology & Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.T.); (K.M.S.); (R.A.); (M.A.C.)
| | - Marion A. Cooley
- Oral Biology & Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.T.); (K.M.S.); (R.A.); (M.A.C.)
| | - Jan M. Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Ahmed A. Elmarakby
- Oral Biology & Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.T.); (K.M.S.); (R.A.); (M.A.C.)
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
4
|
Zhang Z, He Z, Pan J, Yuan M, Lang Y, Wei X, Zhang C. The interaction of BDNF with estrogen in the development of hypertension and obesity, particularly during menopause. Front Endocrinol (Lausanne) 2024; 15:1384159. [PMID: 39655343 PMCID: PMC11625588 DOI: 10.3389/fendo.2024.1384159] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
The expression of BDNF in both neuronal and non-neuronal cells is influenced by various stimuli, including prenatal developmental factors and postnatal conditions such as estrogens, dietary habits, and lifestyle factors like obesity, blood pressure, and aging. Central BDNF plays a crucial role in modulating how target tissues respond to these stimuli, influencing the pathogenesis of hypertension, mitigating obesity, and protecting neurons from aging. Thus, BDNF serves as a dynamic mediator of environmental influences, reflecting an individual's unique history of exposure. Estrogens, on the other hand, regulate various processes to maintain overall physiological well-being. Through nuclear estrogen receptors (ERα, ERβ) and the membrane estrogen receptor (GPER1), estrogens modulate transcriptional processes and signaling events that regulate the expression of target genes, such as ERα, components of the renin-angiotensin system (RAS), and hormone-sensitive lipase. Estrogens are instrumental in maintaining the set point for blood pressure and energy balance. BDNF and estrogens work cooperatively to prevent obesity by favoring lipolysis, and counteractively regulate blood pressure to adapt to the environment. Estrogen deficiency leads to menopause in women with low central BDNF level. This review delves into the complex mechanisms involving BDNF and estrogen, especially in the context of hypertension and obesity, particularly among postmenopausal women. The insights gained aim to inform the development of comprehensive therapeutic strategies for these prevalent syndromes affecting approximately 68% of adults.
Collapse
Affiliation(s)
- Zhongming Zhang
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
- School of Medicine, Zhengzhou University of Industrial Technology, Xinzheng, Henan, China
| | - Ziyi He
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Jing Pan
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Minghui Yuan
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Yini Lang
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Xiaomeng Wei
- School of Medicine, Zhengzhou University of Industrial Technology, Xinzheng, Henan, China
| | - Chaoyun Zhang
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| |
Collapse
|
5
|
Elgazzaz M, Filipeanu C, Lazartigues E. Angiotensin-Converting Enzyme 2 Posttranslational Modifications and Implications for Hypertension and SARS-CoV-2: 2023 Lewis K. Dahl Memorial Lecture. Hypertension 2024; 81:1438-1449. [PMID: 38567498 PMCID: PMC11168885 DOI: 10.1161/hypertensionaha.124.22067] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
ACE2 (angiotensin-converting enzyme 2), a multifunctional transmembrane protein, is well recognized as an important member of the (RAS) renin-angiotensin system with important roles in the regulation of cardiovascular function by opposing the harmful effects of Ang-II (angiotensin II) and AT1R (Ang-II type 1 receptor) activation. More recently, ACE2 was found to be the entry point for the SARS-CoV-2 virus into cells, causing COVID-19. This finding has led to an exponential rise in the number of publications focused on ACE2, albeit these studies often have opposite objectives to the preservation of ACE2 in cardiovascular regulation. However, notwithstanding accumulating data of the role of ACE2 in the generation of angiotensin-(1-7) and SARS-CoV-2 internalization, numerous other putative roles of this enzyme remain less investigated and not yet characterized. Currently, no drug modulating ACE2 function or expression is available in the clinic, and the development of new pharmacological tools should attempt targeting each step of the lifespan of the protein from synthesis to degradation. The present review expands on our presentation during the 2023 Lewis K. Dahl Memorial Lecture Sponsored by the American Heart Association Council on Hypertension. We provide a critical summary of the current knowledge of the mechanisms controlling ACE2 internalization and intracellular trafficking, the mutual regulation with GPCRs (G-protein-coupled receptors) and other proteins, and posttranslational modifications. A major focus is on ubiquitination which has become a critical step in the modulation of ACE2 cellular levels.
Collapse
Affiliation(s)
- Mona Elgazzaz
- Department of Physiology, Augusta University, Medical College of Georgia, Augusta, GA 30912, USA
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Catalin Filipeanu
- Department of Pharmacology, Howard University, Washington, DC 20059, USA
| | - Eric Lazartigues
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA
| |
Collapse
|
6
|
Pan M, Yu M, Zheng S, Luo L, Zhang J, Wu J. Genetic variations in ACE2 gene associated with metabolic syndrome in southern China: a case-control study. Sci Rep 2024; 14:10505. [PMID: 38714718 PMCID: PMC11076479 DOI: 10.1038/s41598-024-61254-5] [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: 11/15/2023] [Accepted: 05/03/2024] [Indexed: 05/10/2024] Open
Abstract
Metabolic syndrome (MetS) is closely related to cardiovascular and cerebrovascular diseases, and genetic predisposition is one of the main triggers for its development. To identify the susceptibility genes for MetS, we investigated the relationship between angiotensin-converting enzyme 2 (ACE2) single nucleotide polymorphisms (SNPs) and MetS in southern China. In total, 339 MetS patients and 398 non-MetS hospitalized patients were recruited. Four ACE2 polymorphisms (rs2074192, rs2106809, rs879922, and rs4646155) were genotyped using the polymerase chain reaction-ligase detection method and tested for their potential association with MetS and its related components. ACE2 rs2074192 and rs2106809 minor alleles conferred 2.485-fold and 3.313-fold greater risks of MetS in women. ACE2 rs2074192 and rs2106809 variants were risk factors for obesity, diabetes, and low-high-density lipoprotein cholesterolemia. However, in men, the ACE2 rs2074192 minor allele was associated with an approximately 0.525-fold reduction in MetS prevalence. Further comparing the components of MetS, ACE2 rs2074192 and rs2106809 variants reduced the risk of obesity and high triglyceride levels. In conclusion, ACE2 rs2074192 and rs2106809 SNPs were independently associated with MetS in a southern Chinese population and showed gender heterogeneity, which can be partially explained by obesity. Thus, these SNPs may be utilized as predictive biomarkers and molecular targets for MetS. A limitation of this study is that environmental and lifestyle differences, as well as genetic heterogeneity among different populations, were not considered in the analysis.
Collapse
Affiliation(s)
- Min Pan
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Fujian Hypertension Research Institute, Fuzhou, 350005, Fujian, People's Republic of China
| | - Mingzhong Yu
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Fujian Hypertension Research Institute, Fuzhou, 350005, Fujian, People's Republic of China
| | - Suli Zheng
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Fujian Hypertension Research Institute, Fuzhou, 350005, Fujian, People's Republic of China
| | - Li Luo
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
- Fujian Hypertension Research Institute, Fuzhou, 350005, Fujian, People's Republic of China
| | - Jie Zhang
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China.
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China.
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China.
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China.
- Fujian Hypertension Research Institute, Fuzhou, 350005, Fujian, People's Republic of China.
| | - Jianmin Wu
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China.
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China.
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China.
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China.
- Fujian Hypertension Research Institute, Fuzhou, 350005, Fujian, People's Republic of China.
| |
Collapse
|
7
|
Liu M, Lu B, Li Y, Yuan S, Zhuang Z, Li G, Wang D, Ma L, Zhu J, Zhao J, Chan CCS, Poon VKM, Chik KKH, Zhao Z, Xian H, Zhao J, Zhao J, Chan JFW, Zhang Y. P21-activated kinase 1 (PAK1)-mediated cytoskeleton rearrangement promotes SARS-CoV-2 entry and ACE2 autophagic degradation. Signal Transduct Target Ther 2023; 8:385. [PMID: 37806990 PMCID: PMC10560660 DOI: 10.1038/s41392-023-01631-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/21/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has had a significant impact on healthcare systems and economies worldwide. The continuous emergence of new viral strains presents a major challenge in the development of effective antiviral agents. Strategies that possess broad-spectrum antiviral activities are desirable to control SARS-CoV-2 infection. ACE2, an angiotensin-containing enzyme that prevents the overactivation of the renin angiotensin system, is the receptor for SARS-CoV-2. ACE2 interacts with the spike protein and facilitates viral attachment and entry into host cells. Yet, SARS-CoV-2 infection also promotes ACE2 degradation. Whether restoring ACE2 surface expression has an impact on SARS-CoV-2 infection is yet to be determined. Here, we show that the ACE2-spike complex is endocytosed and degraded via autophagy in a manner that depends on clathrin-mediated endocytosis and PAK1-mediated cytoskeleton rearrangement. In contrast, free cellular spike protein is selectively cleaved into S1 and S2 subunits in a lysosomal-dependent manner. Importantly, we show that the pan-PAK inhibitor FRAX-486 restores ACE2 surface expression and suppresses infection by different SARS-CoV-2 strains. FRAX-486-treated Syrian hamsters exhibit significantly decreased lung viral load and alleviated pulmonary inflammation compared with untreated hamsters. In summary, our findings have identified novel pathways regulating viral entry, as well as therapeutic targets and candidate compounds for controlling the emerging strains of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ming Liu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 510623, Guangzhou, Guangdong, China
| | - Bingtai Lu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, China
| | - Yue Li
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 510623, Guangzhou, Guangdong, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Zhen Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guangyu Li
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 510623, Guangzhou, Guangdong, China
| | - Dong Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liuheyi Ma
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 510623, Guangzhou, Guangdong, China
| | - Jianheng Zhu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 510623, Guangzhou, Guangdong, China
| | - Jinglu Zhao
- The Third Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Chris Chung-Sing Chan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Vincent Kwok-Man Poon
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Kenn Ka-Heng Chik
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Zhiyao Zhao
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 510623, Guangzhou, Guangdong, China
| | - Huifang Xian
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 510623, Guangzhou, Guangdong, China
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China.
- Guangzhou Laboratory, Guangzhou, Guangdong Province, China.
| | - Yuxia Zhang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 510623, Guangzhou, Guangdong, China.
- The Third Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
8
|
Ghimire B, Pour SK, Middleton E, Campbell RA, Nies MA, Aghazadeh-Habashi A. Renin-Angiotensin System Components and Arachidonic Acid Metabolites as Biomarkers of COVID-19. Biomedicines 2023; 11:2118. [PMID: 37626615 PMCID: PMC10452267 DOI: 10.3390/biomedicines11082118] [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: 06/14/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Through the ACE2, a main enzyme of the renin-angiotensin system (RAS), SARS-CoV-2 gains access into the cell, resulting in different complications which may extend beyond the RAS and impact the Arachidonic Acid (ArA) pathway. The contribution of the RAS through ArA pathways metabolites in the pathogenesis of COVID-19 is unknown. We investigated whether RAS components and ArA metabolites can be considered biomarkers of COVID-19. We measured the plasma levels of RAS and ArA metabolites using an LC-MS/MS. Results indicate that Ang 1-7 levels were significantly lower, whereas Ang II levels were higher in the COVID-19 patients than in healthy control individuals. The ratio of Ang 1-7/Ang II as an indicator of the RAS classical and protective arms balance was dramatically lower in COVID-19 patients. There was no significant increase in inflammatory 19-HETE and 20-HETE levels. The concentration of EETs was significantly increased in COVID-19 patients, whereas the DHETs concentration was repressed. Their plasma levels were correlated with Ang II concentration in COVID-19 patients. In conclusion, evaluating the RAS and ArA pathway biomarkers could provide helpful information for the early detection of high-risk groups, avoid delayed medical attention, facilitate resource allocation, and improve patient clinical outcomes to prevent long COVID incidence.
Collapse
Affiliation(s)
- Biwash Ghimire
- College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (B.G.)
| | - Sana Khajeh Pour
- College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (B.G.)
| | - Elizabeth Middleton
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Robert A. Campbell
- Department of Internal Medicine, Division ofHematology, University of Utah, Salt Lake City, UT 84112, USA
| | - Mary A. Nies
- College of Health, School of Nursing, Idaho State University, Pocatello, ID 83209, USA
| | | |
Collapse
|
9
|
Kataoka H, Nitta K, Hoshino J. Glomerular hyperfiltration and hypertrophy: an evaluation of maximum values in pathological indicators to discriminate "diseased" from "normal". Front Med (Lausanne) 2023; 10:1179834. [PMID: 37521339 PMCID: PMC10372422 DOI: 10.3389/fmed.2023.1179834] [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: 03/05/2023] [Accepted: 06/05/2023] [Indexed: 08/01/2023] Open
Abstract
The success of sodium-glucose cotransporter 2 inhibitors and bariatric surgery in patients with chronic kidney disease has highlighted the importance of glomerular hyperfiltration and hypertrophy in the progression of kidney disease. Sustained glomerular hyperfiltration and hypertrophy can lead to glomerular injury and progressive kidney damage. This article explores the relationship between obesity and chronic kidney disease, focusing on the roles of glomerular hyperfiltration and hypertrophy as hallmarks of obesity-related kidney disease. The pathological mechanisms underlying this association include adipose tissue inflammation, dyslipidemia, insulin resistance, chronic systemic inflammation, oxidative stress, and overactivation of the sympathetic nervous system, as well as the renin-angiotensin aldosterone system. This article explains how glomerular hyperfiltration results from increased renal blood flow and intraglomerular hypertension, inducing mechanical stress on the filtration barrier and post-filtration structures. Injured glomeruli increase in size before sclerosing and collapsing. Therefore, using extreme values, such as the maximal glomerular diameter, could improve the understanding of the data distribution and allow for better kidney failure predictions. This review provides important insights into the mechanisms underlying glomerular hyperfiltration and hypertrophy and highlights the need for further research using glomerular size, including maximum glomerular profile, calculated using needle biopsy specimens.
Collapse
|
10
|
Garrett RE, Palacio CH, Bar-Or D. Long COVID: Is there a kidney link? Front Med (Lausanne) 2023; 10:1138644. [PMID: 37077670 PMCID: PMC10106649 DOI: 10.3389/fmed.2023.1138644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/15/2023] [Indexed: 04/05/2023] Open
Abstract
Metabolic causes such as altered bioenergetics and amino acid metabolism may play a major role in Long COVID. Renal-metabolic regulation is an integral part of these pathways but has not been systematically or routinely investigated in Long COVID. Here we discuss the biochemistry of renal tubular injury as it may contribute to Long COVID symptoms. We propose three potential mechanisms that could be involved in Long COVID namely creatine phosphate metabolism, un-reclaimed glomerular filtrate and COVID specific proximal tubule cells (PTC) injury-a tryptophan paradigm. This approach is intended to allow for improved diagnostics and therapy for the long-haul sufferers.
Collapse
Affiliation(s)
- Raymond E. Garrett
- Swedish Medical Center, Trauma Research Dept., Englewood, CO, United States
| | - Carlos H. Palacio
- South Texas Health System, Trauma Research Dept., McAllen, TX, United States
| | - David Bar-Or
- Swedish Medical Center, Trauma Research Dept., Englewood, CO, United States
- South Texas Health System, Trauma Research Dept., McAllen, TX, United States
- *Correspondence: David Bar-Or,
| |
Collapse
|
11
|
Lott N, Gebhard CE, Bengs S, Haider A, Kuster GM, Regitz-Zagrosek V, Gebhard C. Sex hormones in SARS-CoV-2 susceptibility: key players or confounders? Nat Rev Endocrinol 2023; 19:217-231. [PMID: 36494595 PMCID: PMC9734735 DOI: 10.1038/s41574-022-00780-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2022] [Indexed: 12/14/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has a clear sex disparity in clinical outcomes. Hence, the interaction between sex hormones, virus entry receptors and immune responses has attracted major interest as a target for the prevention and treatment of SARS-CoV-2 infections. This Review summarizes the current understanding of the roles of androgens, oestrogens and progesterone in the regulation of virus entry receptors and disease progression of coronavirus disease 2019 (COVID-19) as well as their therapeutic value. Although many experimental and clinical studies have analysed potential mechanisms by which female sex hormones might provide protection against SARS-CoV-2 infectivity, there is currently no clear evidence for a sex-specific expression of virus entry receptors. In addition, reports describing an influence of oestrogen, progesterone and androgens on the course of COVID-19 vary widely. Current data also do not support the administration of oestradiol in COVID-19. The conflicting evidence and lack of consensus results from a paucity of mechanistic studies and clinical trials reporting sex-disaggregated data. Further, the influence of variables beyond biological factors (sex), such as sociocultural factors (gender), on COVID-19 manifestations has not been investigated. Future research will have to fill this knowledge gap as the influence of sex and gender on COVID-19 will be essential to understanding and managing the long-term consequences of this pandemic.
Collapse
Affiliation(s)
- Nicola Lott
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Achi Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Gabriela M Kuster
- Department of Cardiology and Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Vera Regitz-Zagrosek
- Charité, Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.
- Department of Cardiology, Inselspital Bern University Hospital, Bern, Switzerland.
| |
Collapse
|
12
|
Gaspar AR, Andrade B, Mosca S, Ferreira-Duarte M, Teixeira A, Cosme D, Albino-Teixeira A, Ronchi FA, Leite AP, Casarini DE, Areias JC, Sousa T, Afonso AC, Morato M, Correia-Costa L. Association between blood pressure and angiotensin-converting enzymes activity in prepubertal children ∗. J Hypertens 2023; 41:545-553. [PMID: 36723456 DOI: 10.1097/hjh.0000000000003345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Angiotensin-converting enzymes' (ACEs) relationship with blood pressure (BP) during childhood has not been clearly established. We aimed to compare ACE and ACE2 activities between BMI groups in a sample of prepubertal children, and to characterize the association between these enzymes' activities and BP. METHODS Cross-sectional study of 313 children aged 8-9 years old, included in the birth cohort Generation XXI (Portugal). Anthropometric measurements and 24-h ambulatory BP monitoring were performed. ACE and ACE2 activities were quantified by fluorometric methods. RESULTS Overweight/obese children demonstrated significantly higher ACE and ACE2 activities, when compared to their normal weight counterparts [median (P25-P75), ACE: 39.48 (30.52-48.97) vs. 42.90 (35.62-47.18) vs. 43.38 (33.49-49.89) mU/ml, P for trend = 0.009; ACE2: 10.41 (7.58-15.47) vs. 21.56 (13.34-29.09) vs. 29.00 (22.91-34.32) pM/min per ml, P for trend < 0.001, in normal weight, overweight and obese children, respectively]. In girls, night-time systolic BP (SBP) and diastolic BP (DBP) increased across tertiles of ACE activity ( P < 0.001 and P = 0.002, respectively). ACE2 activity was associated with higher night-time SBP and DBP in overweight/obese girls ( P = 0.037 and P = 0.048, respectively) and night-time DBP in the BMI z-score girl adjusted model ( P = 0.018). Median ACE2 levels were significantly higher among nondipper girls (16.7 vs. 11.6 pM/min per ml, P = 0.009). CONCLUSIONS Our work shows that obesity is associated with activation of the renin-angiotensin-aldosterone system, with significant increase of ACE and ACE2 activities already in childhood. Also, we report sex differences in the association of ACE and ACE2 activities with BP.
Collapse
Affiliation(s)
- Ana R Gaspar
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto
| | - Beatriz Andrade
- Unidade de Saúde Familiar (USF) Nuno Grande, Agrupamento de Centros de Saúde (ACeS) Douro I - Marão e Douro Norte, Vila Real
| | - Sara Mosca
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto
- Department of Pediatrics, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto
| | - Mariana Ferreira-Duarte
- Laboratory of Pharmacology, Department of Drug Sciences
- LAQV@REQUIMTE, Faculdade de Farmácia da Universidade do Porto
| | - Ana Teixeira
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto
- Division of Pediatric Nephrology, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto
| | - Dina Cosme
- Department of Biomedicine - Unit of Pharmacology and Therapeutics, Faculdade de Medicina da Universidade do Porto
- MedInUP - Center for Drug Discovery and Innovative Medicines, Universidade do Porto, Porto
| | - António Albino-Teixeira
- Department of Biomedicine - Unit of Pharmacology and Therapeutics, Faculdade de Medicina da Universidade do Porto
- MedInUP - Center for Drug Discovery and Innovative Medicines, Universidade do Porto, Porto
| | - Fernanda A Ronchi
- Department of Medicine, Nephrology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana P Leite
- Department of Medicine, Nephrology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Dulce E Casarini
- Department of Medicine, Nephrology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José C Areias
- Division of Pediatric Cardiology, Centro Hospitalar Universitário São João
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Teresa Sousa
- Department of Biomedicine - Unit of Pharmacology and Therapeutics, Faculdade de Medicina da Universidade do Porto
- MedInUP - Center for Drug Discovery and Innovative Medicines, Universidade do Porto, Porto
| | - Alberto C Afonso
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto
- Division of Pediatric Nephrology, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto
| | - Manuela Morato
- Laboratory of Pharmacology, Department of Drug Sciences
- LAQV@REQUIMTE, Faculdade de Farmácia da Universidade do Porto
| | - Liane Correia-Costa
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto
- Division of Pediatric Nephrology, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto
| |
Collapse
|
13
|
Sexual Dimorphism in the Expression of Cardiac and Hippocampal Renin-Angiotensin and Kallikrein–Kinin Systems in Offspring from Mice Exposed to Alcohol during Gestation. Antioxidants (Basel) 2023; 12:antiox12030541. [PMID: 36978790 PMCID: PMC10045732 DOI: 10.3390/antiox12030541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Prenatal alcohol exposure (PAE) impairs fetal development. Alcohol consumption was shown to modulate the renin–angiotensin system (RAS). This study aimed to analyze the effects of PAE on the expression of the renin–angiotensin system (RAS) and kallikrein–kinin system (KKS) peptide systems in the hippocampus and heart of mice of both sexes. C57Bl/6 mice were exposed to alcohol during pregnancy at a concentration of 10% (v/v). On postnatal day 45 (PN45), mouse hippocampi and left ventricles (LV) were collected and processed for messenger RNA (mRNA) expression of components of the RAS and KKS. In PAE animals, more pronounced expression of AT1 and ACE mRNAs in males and a restored AT2 mRNA expression in females were observed in both tissues. In LV, increased AT2, ACE2, and B2 mRNA expressions were also observed in PAE females. Furthermore, high levels of H2O2 were observed in males from the PAE group in both tissues. Taken together, our results suggest that modulation of the expression of these peptidergic systems in PAE females may make them less susceptible to the effects of alcohol.
Collapse
|
14
|
Neurology of Systemic Disease. Neurol Clin 2023; 41:399-413. [PMID: 37030966 DOI: 10.1016/j.ncl.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Sex differences exist within the neurologic complications of systemic disease. To promote new avenues for prevention and develop novel therapeutics, we highlight the role of sex in differential outcomes to infectious disease and cardiac arrest and educate the reader in paraneoplastic presentations that may herald underlying malignancies in women.
Collapse
|
15
|
Kataoka H, Nitta K, Hoshino J. Visceral fat and attribute-based medicine in chronic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1097596. [PMID: 36843595 PMCID: PMC9947142 DOI: 10.3389/fendo.2023.1097596] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/13/2023] [Indexed: 02/11/2023] Open
Abstract
Visceral adipose tissue plays a central role in obesity and metabolic syndrome and is an independent risk factor for both cardiovascular and metabolic disorders. Increased visceral adipose tissue promotes adipokine dysregulation and insulin resistance, leading to several health issues, including systemic inflammation, oxidative stress, and activation of the renin-angiotensin-aldosterone system. Moreover, an increase in adipose tissue directly and indirectly affects the kidneys by increasing renal sodium reabsorption, causing glomerular hyperfiltration and hypertrophy, which leads to increased proteinuria and kidney fibrosis/dysfunction. Although the interest in the adverse effects of obesity on renal diseases has grown exponentially in recent years, the relationship between obesity and renal prognosis remains controversial. This may be attributed to the long clinical course of obesity, numerous obesity-related metabolic complications, and patients' attributes. Multiple individual attributes influencing the pathophysiology of fat accumulation make it difficult to understand obesity. In such cases, it may be effective to elucidate the pathophysiology by conducting research tailored to individual attributes from the perspective of attribute-based medicine/personalized medicine. We consider the appropriate use of clinical indicators necessary, according to attributes such as chronic kidney disease stage, level of visceral adipose tissue accumulation, age, and sex. Selecting treatments and clinical indicators based on individual attributes will allow for advancements in the clinical management of patients with obesity and chronic kidney disease. In the clinical setting of obesity-related nephropathy, it is first necessary to accumulate attribute-based studies resulting from the accurate evaluation of visceral fat accumulation to establish evidence for promoting personalized medicine.
Collapse
|
16
|
Estrogen as a key regulator of energy homeostasis and metabolic health. Biomed Pharmacother 2022; 156:113808. [DOI: 10.1016/j.biopha.2022.113808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022] Open
|
17
|
Suzuki K, Hekmatikar AHA, Jalalian S, Abbasi S, Ahmadi E, Kazemi A, Ruhee RT, Khoramipour K. The Potential of Exerkines in Women's COVID-19: A New Idea for a Better and More Accurate Understanding of the Mechanisms behind Physical Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph192315645. [PMID: 36497720 PMCID: PMC9737724 DOI: 10.3390/ijerph192315645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 05/31/2023]
Abstract
The benefits of physical exercise are well-known, but there are still many questions regarding COVID-19. Chow et al.'s 2022 study, titled Exerkines and Disease, showed that a special focus on exerkines can help to better understand the underlying mechanisms of physical exercise and disease. Exerkines are a group of promising molecules that may underlie the beneficial effects of physical exercise in diseases. The idea of exerkines is to understand the effects of physical exercise on diseases better. Exerkines have a high potential for the treatment of diseases and, considering that, there is still no study of the importance of exerkines on the most dangerous disease in the world in recent years, COVID-19. This raises the fundamental question of whether exerkines have the potential to manage COVID-19. Most of the studies focused on the general changes in physical exercise in patients with COVID-19, both during the illness and after discharge from the hospital, and did not investigate the basic differences. A unique look at the management of COVID-19 by exerkines, especially in obese and overweight women who experience high severity of COVID-19 and whose recovery period is long after discharge from the hospital, can help to understand the basic mechanisms. In this review, we explore the potential of exerkines in COVID-19 by practicing physical exercise to provide compelling practice recommendations with new insights.
Collapse
Affiliation(s)
- Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
| | - Amir Hossein Ahmadi Hekmatikar
- Department of Physical Education and Sport Sciences, Faculty of Humanities, Tarbiat Modares University, Tehran 10600, Iran
| | - Shadi Jalalian
- Department of Physical Education and Sport Sciences, Science and Research Branch, Islamic Azad University, Tehran 10600, Iran
| | - Shaghayegh Abbasi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Kharazmi University, Tehran 10600, Iran
| | - Elmira Ahmadi
- Department of Physical Education and Sport Sciences, Faculty of Humanities, Tarbiat Modares University, Tehran 10600, Iran
| | - Abdolreza Kazemi
- Department of Sports Science, Faculty of Literature and Humanities, Vali-e-Asr University, Rafsanjan 7718897111, Iran
| | | | - Kayvan Khoramipour
- Neuroscience Research Center, Institute of Neuropharmacology, Department of Physiology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman 7616914115, Iran
| |
Collapse
|
18
|
Taslem Mourosi J, Anwar S, Hosen MJ. The sex and gender dimensions of COVID-19: A narrative review of the potential underlying factors. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 103:105338. [PMID: 35863677 PMCID: PMC9288935 DOI: 10.1016/j.meegid.2022.105338] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/05/2022] [Accepted: 07/09/2022] [Indexed: 12/17/2022]
Abstract
Multiple lines of evidence indicate that the male sex is a significant risk factor for severe disease and mortality due to coronavirus disease 2019 (COVID-19). However, the precise explanation for the discrepancy is currently unclear. Immunologically, the female-biased protection against COVID-19 could presumably be due to a more rapid and robust immune response to viruses exhibited by males. The female hormones, e.g., estrogens and progesterone, may have protective roles against viral infections. In contrast, male hormones, e.g., testosterone, can act oppositely. Besides, the expression of the ACE-2 receptor in the lung and airway lining, which the SARS-CoV-2 uses to enter cells, is more pronounced in males. Estrogen potentially plays a role in downregulating the expression of ACE-2, which could be a plausible biological explanation for the reduced severity of COVID-19 in females. Comorbidities, e.g., cardiovascular diseases, diabetes, and kidney disorders, are considered significant risk factors for severe outcomes in COVID-19. Age-adjusted data shows that males are statistically more predisposed to these morbidities-amplifying risks for males with COVID-19. In addition, many sociocultural factors and gender-constructed behavior of men and women impact exposure to infections and outcomes. In many parts of the world, women are more likely to abide by health regulations, e.g., mask-wearing and handwashing, than men. In contrast, men, in general, are more involved with high-risk behaviors, e.g., smoking and alcohol consumption, and high-risk jobs that require admixing with people, which increases their risk of exposure to the infection. Overall, males and females suffer differently from COVID-19 due to a complex interplay between many biological and sociocultural factors.
Collapse
Affiliation(s)
- Jarin Taslem Mourosi
- Department of Biology, The Catholic University of America, Washington, DC, USA; Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh; Bacteriophage Medical Research Center, The Catholic University of America, Washington, DC, USA.
| | - Saeed Anwar
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada; Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh.
| | - Mohammad Jakir Hosen
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh.
| |
Collapse
|
19
|
RACHDI R, HANNACHI S, ZRIBI S, AYED O, ABID R, MOATEMRI Z, MHAMDI S, DABBOUSSI S, GHARSALLAH H, SELLAMI W, SAMMOUD W, MASSOUDI H, LAMINE K, DJEBBI O, HAMMAMI R, BEN MOUSSA M, BELLAAJ R, BATTIKH R, RACHDI MR, FERJANI M. [Comparative study of patients hospitalized for SARS-CoV-2 infection in two consecutive waves in Tunisia]. MEDECINE TROPICALE ET SANTE INTERNATIONALE 2022; 2:mtsi.v2i3.2022.207. [PMID: 36284562 PMCID: PMC9557822 DOI: 10.48327/mtsi.v2i3.2022.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022]
Abstract
Introduction Since December 2019, a novel coronavirus (SARS-CoV-2) has triggered a global pandemic with a heavy medical and societal-economic toll. The health consequences were not similar during the successive waves that affected several countries. The aim of our study was to compare the sociodemographic, clinical and evolutionary features of COVID-19 patients hospitalized at the Military Hospital of Tunis (HMPIT) during the 2nd and 3rd waves that affected the country. Patients and methods Observational prospective study involving 1,527 COVID-19 patients hospitalized at HMPIT over 11 months, divided into two periods: from July 2020 to December 2020 called the second wave (V2) and from January 2021 to May 2021 called the third wave (V3). We compared the epidemiological data, the clinical form and the evolution of the patients for each period. Results The number of hospitalized patients was 636 during V2 compared to 891 during V3. Average age was 63.5 ± 15.3 years during V2 versus 65.8 ± 17.8 years during V3 (P = not significant [NS]). The percentage of young adults [18-40 years] was 6.5% during V2 compared to 6.7% during V3 (P = NS). The gender ratio (M/F) was 1.59 for V2 and 1.42 for V3 (P = NS). Comorbidities were present in 65% of V2 patients and 66.3% of V3 patients (P = NS), with hypertension being the most prevalent one in both groups (47.2% for V2 versus 44.9% for V3; P = NS), followed by overweight, dyslipidemia and diabetes (33% for V2 versus 39.3% for V3; P = 0.012). The median duration between symptoms onset and hospitalization was 7 days [5-10] during V2 versus 8.5 days during V3 [5-12] (P = 0.0004). The severe clinical form was present in 49% of patients admitted during V2 compared to 34.8% during V3 (P < 10-3). The critical form represented 18.6% of cases during V2 against 16.8% during V3 (P = NS). The average hospital length of stay in COVID units (outside of intensive care unit) was 8.4 ± 5.4 days during V2 and 9.8 ± 5.7 days during V3. The average length of stay was significantly longer for the intensive care unit (11.3 ± 3.4 days for V2 versus 13.8 ± 3.9 days for V3; P = 0.01). The case fatality rate was 24.5% during V2 and 20.7% during V3 (P = NS). Median age of death was 70.2 years [42-88] during V2 and 70.4 years [22-96] during V3 with 2 patients less than 40 years of age (1%) for the latter period. The gender ratio (M/F) of deceased patients was 3.21 for V2 and 1.5 for V3 (P = 0.001). The case fatality rate was higher in the intensive care unit (65.4% for V2 versus 69.7% for V3; P = NS). Causes of death were dominated by ARDS (acute respiratory distress syndrome) for both periods (55.1% for V2 versus 70.8% for V3; P = 0.002), followed by septic shock (12.8% for V2 versus 10.8% for V3; P = NS) and multi-organ failure (9.6% for V2 versus 7.0% for V3; P = NS). Conclusion This study revealed a decrease in severe and critical clinical forms during the 3rd wave, as well as a decrease in the case fatality rate compared to the previous wave, due to improved management and vaccination. On the other hand, the percentage of ARDS was significantly higher during this wave probably related to the beginning of circulation in our country of the Delta variant causing more severe clinical cases.
Collapse
Affiliation(s)
- Rim RACHDI
- Service d'hygiène hospitalière et de protection de l'environnement, Hôpital militaire principal d'instruction de Tunis (HMPIT), 1008 Montfleury, Tunis, Tunisie,*
| | - Souha HANNACHI
- Service des maladies infectieuses, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Sabrine ZRIBI
- Service d'hygiène hospitalière et de protection de l'environnement, Hôpital militaire principal d'instruction de Tunis (HMPIT), 1008 Montfleury, Tunis, Tunisie,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Oumaima AYED
- Service d'hygiène hospitalière et de protection de l'environnement, Hôpital militaire principal d'instruction de Tunis (HMPIT), 1008 Montfleury, Tunis, Tunisie,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Rim ABID
- Service des maladies infectieuses, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Zied MOATEMRI
- Service de pneumo-phtisiologie, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Samira MHAMDI
- Service de pneumo-phtisiologie, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Salsabil DABBOUSSI
- Service de pneumo-phtisiologie, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Hédi GHARSALLAH
- Service d'anesthésie-réanimation, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Walid SELLAMI
- Service d'anesthésie-réanimation, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | | | - Hakim MASSOUDI
- Service d'anesthésie-réanimation, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Khaled LAMINE
- Service d'accueil des urgences, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Olfa DJEBBI
- Service d'accueil des urgences, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Rim HAMMAMI
- Service d'accueil des urgences, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | | | - Ridha BELLAAJ
- Service d'hygiène hospitalière et de protection de l'environnement, Hôpital militaire principal d'instruction de Tunis (HMPIT), 1008 Montfleury, Tunis, Tunisie,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Riadh BATTIKH
- Service des maladies infectieuses, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Mohamed Radhouane RACHDI
- Service de gynécologie obstétrique, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| | - Mustapha FERJANI
- Service d'anesthésie-réanimation, HMPIT,Faculté de médecine de Tunis, Université Tunis El Manar, Tunisie
| |
Collapse
|
20
|
Nazari S, Moosavi SMS. Temporal patterns of alterations in obesity index, lipid profile, renal function and blood pressure during the development of hypertension in male, but not female, rats fed a moderately high-fat diet. Arch Physiol Biochem 2022; 128:897-909. [PMID: 32195603 DOI: 10.1080/13813455.2020.1739713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Male Sprague-Dawley rats consuming a moderately high-fat (MHF)-diet diverge into obesity-prone (OP) with hypertension and obesity-resistant. OBJECTIVES To study the temporal inter-relationships between body-weight, obesity-index, plasma lipid-profile, renal functional parameters and systolic-pressure alterations during 10-weeks feeding MHF or normal diet to male and female rats. METHODS Body-weight, obesity-index and systolic-pressure were measured weekly, while metabolic-cage and blood-sampling protocols were performed every other week. After 10-weeks, renal excretory responses to acute salt-loading and renal autoregulation were examined. RESULTS The male-OP group had progressively increased body-weight, plasma-triglyceride and systolic-pressure from Weeks 2, 4 and 5, respectively, lower renal sodium-excretion at weeks 4-8 and finally, delayed excretory response to salt-loading and rightward and downward shifts in renal autoregulatory curves compared to all other groups. CONCLUSION Feeding the MHF-diet in male-OP rats led to a greater weight-gain and adiposity followed by the development of atherogenic-hyperlipidaemia and persistently impaired pressure-natriuresis to induce hypertension.
Collapse
Affiliation(s)
- Somayeh Nazari
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mostafa Shid Moosavi
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
21
|
Herichová I, Jendrisková S, Pidíková P, Kršková L, Olexová L, Morová M, Stebelová K, Štefánik P. Effect of 17β-estradiol on the daily pattern of ACE2, ADAM17, TMPRSS2 and estradiol receptor transcription in the lungs and colon of male rats. PLoS One 2022; 17:e0270609. [PMID: 35763527 PMCID: PMC9239479 DOI: 10.1371/journal.pone.0270609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022] Open
Abstract
Covid-19 progression shows sex-dependent features. It is hypothesized that a better Covid-19 survival rate in females can be attributed to the presence of higher 17β-estradiol (E2) levels in women than in men. Virus SARS-CoV-2 is enabled to enter the cell with the use of angiotensin converting enzyme 2 (ACE2). The expression of several renin-angiotensin system components has been shown to exert a rhythmic pattern, and a role of the circadian system in their regulation has been implicated. Therefore, the aim of the study is to elucidate possible interference between E2 signalling and the circadian system in the regulation of the expression of ACE2 mRNA and functionally related molecules. E2 was administered at a dosage of 40 μg/kg/day for 7 days to male Wistar rats, and sampling of the lungs and colon was performed during a 24-h cycle. The daily pattern of expression of molecules facilitating SARS-CoV-2 entry into the cell, clock genes and E2 receptors was analysed. As a consequence of E2 administration, a rhythm in ACE2 and TMPRSS2 mRNA expression was observed in the lungs but not in the colon. ADAM17 mRNA expression showed a pronounced rhythmic pattern in both tissues that was not influenced by E2 treatment. ESR1 mRNA expression exerted a rhythmic pattern, which was diminished by E2 treatment. The influence of E2 administration on ESR2 and GPER1 mRNA expression was greater in the lungs than in the colon as a significant rhythm in ESR2 and GPER1 mRNA expression appeared only in the lungs after E2 treatment. E2 administration also increased the amplitude of bmal1 expression in the lungs, which implicates altered functioning of peripheral oscillators in response to E2 treatment. The daily pattern of components of the SARS-CoV-2 entrance pathway and their responsiveness to E2 should be considered in the timing of pharmacological therapy for Covid-19.
Collapse
Affiliation(s)
- Iveta Herichová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Soňa Jendrisková
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Paulína Pidíková
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Lucia Kršková
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Lucia Olexová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Martina Morová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Katarína Stebelová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Peter Štefánik
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| |
Collapse
|
22
|
Detection of SARS-CoV-2 in subcutaneous fat but not visceral fat, and the disruption of fat lymphocyte homeostasis in both fat tissues in the macaque. Commun Biol 2022; 5:542. [PMID: 35661814 PMCID: PMC9166782 DOI: 10.1038/s42003-022-03503-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 05/19/2022] [Indexed: 12/13/2022] Open
Abstract
The well documented association between obesity and the severity of SARS-CoV-2 infection raises the question of whether adipose tissue (AT) is impacted during this infection. Using a model of SARS-CoV-2 infection in cynomolgus macaques, we detected the virus within subcutaneous AT (SCAT) but not in visceral AT (VAT) or epicardial AT on day 7 post-infection. We sought to determine the mechanisms responsible for this selective detection and observed higher levels of angiotensin-converting-enzyme-2 mRNA expression in SCAT than in VAT. Lastly, we evaluated the immunological consequences of SARS-CoV-2 infection on AT: both SCAT and VAT T cells showed a drastic reduction in CD69 expression, a standard marker of resident memory T cell in tissue, that is also involved in the migratory and metabolic properties of T cells. Our results demonstrate that in a model of mild infection, SCAT is selectively infected by SARS-CoV-2 although changes in the immune properties of AT are observed in both SCAT and VAT. Subcutaneous fat tissue expresses higher angiotensin-converting-enzyme 2 mRNA than visceral fat tissue and is selectively infected by SARS-Cov-2, while both fat tissues show drastic reduction in CD69 expression in T cells.
Collapse
|
23
|
Shamoon L, Romero A, De la Cuesta F, Sánchez-Ferrer CF, Peiró C. Angiotensin-(1-7), a protective peptide against vascular aging. Peptides 2022; 152:170775. [PMID: 35231551 DOI: 10.1016/j.peptides.2022.170775] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/09/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022]
Abstract
Vascular aging is a complex and multifaceted process that provokes profound molecular, structural, and functional changes in the vasculature. Eventually, these profound aging alterations make arteries more prone to vascular disease, including hypertension, atherosclerosis and other arterial complications that impact the organism beyond the cardiovascular system and accelerate frailty. For these reasons, preventing or delaying the hallmarks of vascular aging is nowadays a major health goal, especially in our aged societies. In this context, angiotensin(Ang)-(1-7), a major player of the protective branch of the renin-angiotensin system, has gained relevance over recent years as growing knowledge on its anti-aging properties is being unveiled. Here, we briefly review the main actions of Ang-(1-7) against vascular aging. These include protection against vascular cell senescence, anti-inflammatory and antioxidant effects together with the induction of cytoprotective systems. Ang-(1-7) further ameliorates endothelial dysfunction, a hallmark of vascular aging and disease, attenuates fibrosis and calcification and promotes protective angiogenesis and repair. Although further research is needed to better understand the anti-aging properties of Ang-(1-7) on the vasculature, this heptapeptide arises as a promising pharmacological tool for preventing vascular aging and frailty.
Collapse
Affiliation(s)
- L Shamoon
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Paz, IdIPAZ, Madrid, Spain
| | - A Romero
- German Center for the Study of Diabetes, Düsseldorf, Germany
| | - F De la Cuesta
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Spain.
| | - C F Sánchez-Ferrer
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Paz, IdIPAZ, Madrid, Spain.
| | - C Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Paz, IdIPAZ, Madrid, Spain.
| |
Collapse
|
24
|
Damen MSMA, Alarcon PC, Shah AS, Divanovic S. Greasing the inflammatory pathogenesis of viral pneumonias in diabetes. Obes Rev 2022; 23:e13415. [PMID: 34989117 PMCID: PMC9771603 DOI: 10.1111/obr.13415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/16/2022]
Abstract
Type 2 diabetes (T2D) and obesity are independent risk factors for increased morbidity and mortality associated with influenza and SARS-CoV-2 infection. Skewed cellular metabolism shapes immune cell inflammatory responsiveness and function in obesity, T2D, and infection. However, altered immune cell responsiveness and levels of systemic proinflammatory mediators, partly independent of peripheral immune cell contribution, are linked with SARS-CoV-2-associated disease severity. Despite such knowledge, the role of tissue parenchymal cell-driven inflammatory responses, and specifically those dominantly modified in obesity (e.g., adipocytes), in influenza and SARS-CoV-2 infection pathogenesis remain poorly defined. Whether obesity-dependent skewing of adipocyte cellular metabolism uncovers inflammatory clades and promotes the existence of a 'pathogenic-inflammatory' adipocyte phenotype that amplifies SARS-CoV-2 infection diseases severity in individuals with obesity and individuals with obesity and T2D has not been examined. Here, using the knowledge gained from studies of immune cell responses in obesity, T2D, and infection, we highlight the key knowledge gaps underlying adipocyte cellular functions that may sculpt and grease pathogenic processes associated with influenza and SARS-CoV-2 disease severity in diabetes.
Collapse
Affiliation(s)
- Michelle S M A Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Pablo C Alarcon
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, Cincinnati Childrens Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Amy S Shah
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, Cincinnati Childrens Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
25
|
Foolchand A, Ghazi T, Chuturgoon AA. Malnutrition and Dietary Habits Alter the Immune System Which May Consequently Influence SARS-CoV-2 Virulence: A Review. Int J Mol Sci 2022; 23:2654. [PMID: 35269795 PMCID: PMC8910702 DOI: 10.3390/ijms23052654] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19, resulting from the SARS-CoV-2 virus, is a major pandemic that the world is fighting. SARS-CoV-2 primarily causes lung infection by attaching to the ACE2 receptor on the alveolar epithelial cells. However, the ACE2 receptor is also present in intestinal epithelial cells, suggesting a link between nutrition, virulence and clinical outcomes of COVID-19. Respiratory viral infections perturb the gut microbiota. The gut microbiota is shaped by our diet; therefore, a healthy gut is important for optimal metabolism, immunology and protection of the host. Malnutrition causes diverse changes in the immune system by repressing immune responses and enhancing viral vulnerability. Thus, improving gut health with a high-quality, nutrient-filled diet will improve immunity against infections and diseases. This review emphasizes the significance of dietary choices and its subsequent effects on the immune system, which may potentially impact SARS-CoV-2 vulnerability.
Collapse
Affiliation(s)
| | | | - Anil A. Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Sciences, Howard College Campus, University of Kwa-Zulu Natal, Durban 4041, South Africa; (A.F.); (T.G.)
| |
Collapse
|
26
|
Liu A, Raja xavier J, Singh Y, Brucker SY, Salker MS. Molecular and Physiological Aspects of SARS-CoV-2 Infection in Women and Pregnancy. Front Glob Womens Health 2022; 3:756362. [PMID: 35284910 PMCID: PMC8908006 DOI: 10.3389/fgwh.2022.756362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 02/01/2022] [Indexed: 01/08/2023] Open
Abstract
Whilst scientific knowledge about SARS-CoV-2 and COVID-19 is rapidly increasing, much of the effects on pregnant women is still unknown. To accommodate pregnancy, the human endometrium must undergo a physiological transformation called decidualization. These changes encompass the remodeling of endometrial immune cells leading to immunotolerance of the semi-allogenic conceptus as well as defense against pathogens. The angiotensin converting enzyme 2 (ACE2) plays an important regulatory role in the renin-angiotensin-system (RAS) and has been shown to be protective against comorbidities known to worsen COVID-19 outcomes. Furthermore, ACE2 is also crucial for decidualization and thus for early gestation. An astounding gender difference has been found in COVID-19 with male patients presenting with more severe cases and higher mortality rates. This could be attributed to differences in sex chromosomes, hormone levels and behavior patterns. Despite profound changes in the female body during pregnancy, expectant mothers do not face worse outcomes compared with non-pregnant women. Whereas mother-to-child transmission through respiratory droplets during labor or in the postnatal period is known, another question of in utero transmission remains unanswered. Evidence of placental SARS-CoV-2 infection and expression of viral entry receptors at the maternal-fetal interface suggests the possibility of in utero transmission. SARS-CoV-2 can cause further harm through placental damage, maternal systemic inflammation, and hindered access to health care during the pandemic. More research on the effects of COVID-19 during early pregnancy as well as vaccination and treatment options for gravid patients is urgently needed.
Collapse
Affiliation(s)
- Anna Liu
- Research Institute of Women's Health, Eberhard Karls University, Tübingen, Germany
| | - Janet Raja xavier
- Research Institute of Women's Health, Eberhard Karls University, Tübingen, Germany
| | - Yogesh Singh
- Research Institute of Women's Health, Eberhard Karls University, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls University, Tübingen, Germany
| | - Sara Y. Brucker
- Research Institute of Women's Health, Eberhard Karls University, Tübingen, Germany
| | - Madhuri S. Salker
- Research Institute of Women's Health, Eberhard Karls University, Tübingen, Germany
| |
Collapse
|
27
|
Mechanisms contributing to adverse outcomes of COVID-19 in obesity. Mol Cell Biochem 2022; 477:1155-1193. [PMID: 35084674 PMCID: PMC8793096 DOI: 10.1007/s11010-022-04356-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/07/2022] [Indexed: 01/08/2023]
Abstract
A growing amount of epidemiological data from multiple countries indicate an increased prevalence of obesity, more importantly central obesity, among hospitalized subjects with COVID-19. This suggests that obesity is a major factor contributing to adverse outcome of the disease. As it is a metabolic disorder with dysregulated immune and endocrine function, it is logical that dysfunctional metabolism contributes to the mechanisms behind obesity being a risk factor for adverse outcome in COVID-19. Emerging data suggest that in obese subjects, (a) the molecular mechanisms of viral entry and spread mediated through ACE2 receptor, a multifunctional host cell protein which links to cellular homeostasis mechanisms, are affected. This includes perturbation of the physiological renin-angiotensin system pathway causing pro-inflammatory and pro-thrombotic challenges (b) existent metabolic overload and ER stress-induced UPR pathway make obese subjects vulnerable to severe COVID-19, (c) host cell response is altered involving reprogramming of metabolism and epigenetic mechanisms involving microRNAs in line with changes in obesity, and (d) adiposopathy with altered endocrine, adipokine, and cytokine profile contributes to altered immune cell metabolism, systemic inflammation, and vascular endothelial dysfunction, exacerbating COVID-19 pathology. In this review, we have examined the available literature on the underlying mechanisms contributing to obesity being a risk for adverse outcome in COVID-19.
Collapse
|
28
|
Xue B, Yu Y, Beltz TG, Guo F, Wei SG, Johnson AK. Loss of the Protective Effect of Estrogen Contributes to Maternal Gestational Hypertension-Induced Hypertensive Response Sensitization Elicited by Postweaning High-Fat Diet in Female Offspring. J Am Heart Assoc 2022; 11:e023685. [PMID: 35014859 PMCID: PMC9238517 DOI: 10.1161/jaha.121.023685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background A recent study conducted in male offspring demonstrated that maternal gestational hypertension (MHT) induces hypertensive response sensitization (HTRS) elicited by postweaning high‐fat diet (HFD). In this study, we investigated the sensitizing effect of MHT on postweaning HFD‐induced hypertensive response in female rat offspring and assessed the protective role of estrogen in HTRS. Methods and Results The results showed that MHT also induced a sensitized HFD‐elicited hypertensive response in intact female offspring. However, compared with male offspring, this MHT‐induced HTRS was sex specific in that intact female offspring exhibited an attenuated increase in blood pressure. Ovariectomy significantly enhanced the HFD‐induced increase in blood pressure and the pressor response to centrally administered angiotensin II or tumor necrosis factor‐α in offspring of normotensive dams, which was accompanied by elevated centrally driven sympathetic activity, upregulated mRNA expression of prohypertensive components, and downregulated expression of antihypertensive components in the hypothalamic paraventricular nucleus. However, when compared with HFD‐fed ovariectomized offspring of normotensive dams, the MHT‐induced HTRS and pressor responses to centrally administered angiotensin II or tumor necrosis factor‐α in HFD‐fed intact offspring of MHT dams were not potentiated by ovariectomy, but the blood pressure and elicited pressor responses as well as central sympathetic tone remained higher. Conclusions The results indicate that in adult female offspring MHT induced HTRS elicited by HFD. Estrogen normally plays a protective role in antagonizing HFD prohypertensive effects, and MHT compromises this normal protective action of estrogen by augmenting brain reactivity and centrally driven sympathetic activity.
Collapse
Affiliation(s)
- Baojian Xue
- Department of Psychological and Brain Sciences University of Iowa Iowa City IA
| | - Yang Yu
- Department of Internal Medicine University of Iowa Iowa City IA
| | - Terry G Beltz
- Department of Psychological and Brain Sciences University of Iowa Iowa City IA
| | - Fang Guo
- Department of Psychological and Brain Sciences University of Iowa Iowa City IA
| | - Shun-Guang Wei
- Department of Internal Medicine University of Iowa Iowa City IA.,François M. Abboud Cardiovascular Research Center University of Iowa Iowa City IA
| | - Alan Kim Johnson
- Department of Psychological and Brain Sciences University of Iowa Iowa City IA.,Department of Neuroscience and Pharmacology University of Iowa Iowa City IA.,Department of Health and Human Physiology University of Iowa Iowa City IA.,François M. Abboud Cardiovascular Research Center University of Iowa Iowa City IA
| |
Collapse
|
29
|
Leach DA, Brooke GN, Bevan CL. Roles of steroid receptors in the lung and COVID-19. Essays Biochem 2021; 65:1025-1038. [PMID: 34328182 PMCID: PMC8628186 DOI: 10.1042/ebc20210005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/13/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022]
Abstract
COVID-19 symptoms and mortality are largely due to its devastating effects in the lungs. The disease is caused by the SARS (Severe Acute Respiratory Syndrome)-CoV-2 coronavirus, which requires host cell proteins such as ACE2 (angiotensin-converting enzyme 2) and TMPRSS2 (transmembrane serine protease 2) for infection of lung epithelia. The expression and function of the steroid hormone receptor family is important in many aspects that impact on COVID-19 effects in the lung - notably lung development and function, the immune system, and expression of TMPRSS2 and ACE2. This review provides a brief summary of current knowledge on the roles of the steroid hormone receptors [androgen receptor (AR), glucocorticoid receptor (GR), progesterone receptor (PR), mineralocorticoid receptor (MR) and oestrogen receptor (ER)] in the lung, their effects on host cell proteins that facilitate SARS-CoV-2 uptake, and provides a snapshot of current clinical trials investigating the use of steroid receptor (SR) ligands to treat COVID-19.
Collapse
Affiliation(s)
- Damien A. Leach
- Division of Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, U.K
| | - Greg N. Brooke
- Division of Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, U.K
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, U.K
| | - Charlotte L. Bevan
- Division of Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, U.K
| |
Collapse
|
30
|
Sarzani R, Allevi M, Giulietti F, Di Pentima C, Re S, Giordano P, Spannella F. The Identikit of Patient at Risk for Severe COVID-19 and Death: The Dysregulation of Renin-Angiotensin System as the Common Theme. J Clin Med 2021; 10:5883. [PMID: 34945176 PMCID: PMC8704645 DOI: 10.3390/jcm10245883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/24/2022] Open
Abstract
Since the first months of the coronavirus disease 2019 (COVID-19) pandemic, several specific physiologic traits, such as male sex and older age, or health conditions, such as overweight/obesity, arterial hypertension, metabolic syndrome, and type 2 diabetes mellitus, have been found to be highly prevalent and associated with increased risk of adverse outcomes in hospitalized patients. All these cardiovascular morbidities are widespread in the population and often coexist, thus identifying a common patient phenotype, characterized by a hyper-activation of the "classic" renin-angiotensin system (RAS) and mediated by the binding of angiotensin II (Ang II) to the type 1-receptor. At the same time, the RAS imbalance was proved to be crucial in the genesis of lung injury after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, where angiotensin-converting-enzyme-2 (ACE2) is not only the receptor for SARS-CoV-2, but its down-regulation through internalization and shedding, caused by the virus binding, leads to a further dysregulation of RAS by reducing angiotensin 1-7 (Ang 1-7) production. This focused narrative review will discuss the main available evidence on the role played by cardiovascular and metabolic conditions in severe COVID-19, providing a possible pathophysiological link based on the disequilibrium between the two opposite arms of RAS.
Collapse
Affiliation(s)
- Riccardo Sarzani
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| | - Massimiliano Allevi
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| | - Federico Giulietti
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| | - Chiara Di Pentima
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| | - Serena Re
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| | - Piero Giordano
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
| | - Francesco Spannella
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| |
Collapse
|
31
|
Bengs S, Rossi A, Haberecker M, Mikail N, Meisel A, Haider A, Grämer M, Portmann A, Todorov A, Schönenberger C, Gebhard CE, Kuster GM, Regitz-Zagrosek V, Gebhard C. Immunoreactivity of the SARS-CoV-2 entry proteins ACE-2 and TMPRSS-2 in murine models of hormonal manipulation, ageing, and cardiac injury. Sci Rep 2021; 11:23993. [PMID: 34907257 PMCID: PMC8671541 DOI: 10.1038/s41598-021-03181-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/22/2021] [Indexed: 12/24/2022] Open
Abstract
Previous work indicates that SARS-CoV-2 virus entry proteins angiotensin-converting enzyme 2 (ACE-2) and the cell surface transmembrane protease serine 2 (TMPRSS-2) are regulated by sex hormones. However, clinical studies addressing this association have yielded conflicting results. We sought to analyze the impact of sex hormones, age, and cardiovascular disease on ACE-2 and TMPRSS-2 expression in different mouse models. ACE-2 and TMPRSS-2 expression was analyzed by immunostaining in a variety of tissues obtained from FVB/N mice undergoing either gonadectomy or sham-surgery and being subjected to ischemia-reperfusion injury or transverse aortic constriction surgery. In lung tissues sex did not have a significant impact on the expression of ACE-2 and TMPRSS-2. On the contrary, following myocardial injury, female sex was associated to a lower expression of ACE-2 at the level of the kidney tubules. In addition, after myocardial injury, a significant correlation between younger age and higher expression of both ACE-2 and TMPRSS-2 was observed for lung alveoli and bronchioli, kidney tubules, and liver sinusoids. Our experimental data indicate that gonadal hormones and biological sex do not alter ACE-2 and TMPRSS-2 expression in the respiratory tract in mice, independent of disease state. Thus, sex differences in ACE-2 and TMPRSS-2 protein expression observed in mice may not explain the higher disease burden of COVID-19 among men.
Collapse
Affiliation(s)
- Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alexia Rossi
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Martina Haberecker
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Nidaa Mikail
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alexander Meisel
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Achi Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, and Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Muriel Grämer
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Angela Portmann
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Atanas Todorov
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | | | - Gabriela M Kuster
- Department of Cardiology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, Myocardial Research, University of Basel, Basel, Switzerland
| | - Vera Regitz-Zagrosek
- Charité, Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
32
|
Pedrianes-Martin PB, Martin-Rincon M, Morales-Alamo D, Perez-Suarez I, Perez-Valera M, Galvan-Alvarez V, Curtelin D, de Pablos-Velasco P, Calbet JAL. Treatment of hypertension with angiotensin-converting enzyme inhibitors or angiotensin receptor blockers and resting metabolic rate: A cross-sectional study. J Clin Hypertens (Greenwich) 2021; 23:2106-2114. [PMID: 34846787 PMCID: PMC8696230 DOI: 10.1111/jch.14392] [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: 07/01/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/26/2022]
Abstract
Hypertension in obese and overweight patients is associated with an elevated resting metabolic rate (RMR). The aim of this study was to determine whether RMR is reduced in hypertensive patients treated with angiotensin‐converting enzyme inhibitors (ACEI) and blockers (ARB). The RMR was determined by indirect calorimetry in 174 volunteers; 93 (46.5 %) were hypertensive, of which 16 men and 13 women were treated with ACEI/ARB, while 30 men and 19 women with untreated hypertension served as a control group. Treated and untreated hypertensives had similar age, BMI, physical activity, and cardiorespiratory fitness. The RMR normalized to the lean body mass (LBM) was 15% higher in the untreated than ACEI/ARB‐treated hypertensive women (p = .003). After accounting for LBM, whole‐body fat mass, age, the double product (heart rate x systolic blood pressure), and the distance walked per day, the RMR was 2.9% lower in the patients taking ACEI/ARB (p = .26, treatment x sex interaction p = .005). LBM, age, and the double product explained 78% of the variability in RMR (R2 = 0.78, p < .001). In contrast, fat mass, the distance walked per day, and total T4 or TSH did not add predictive power to the model. Compared to men, a greater RMR per kg of LBM was observed in untreated hypertensive overweight and obese women, while this sex difference was not observed in patients treated with ACEI or ARBs. In conclusion, our results indicate that elevated RMR per kg of LBM may be normalized by antagonizing the renin‐angiotensin system.
Collapse
Affiliation(s)
- Pablo B Pedrianes-Martin
- Department of Endocrinology and Nutrition, Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria, Spain.,Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Marcos Martin-Rincon
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain.,Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - David Morales-Alamo
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain.,Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Ismael Perez-Suarez
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain.,Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Mario Perez-Valera
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain.,Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Victor Galvan-Alvarez
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain.,Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - David Curtelin
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Pedro de Pablos-Velasco
- Department of Endocrinology and Nutrition, Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria, Spain.,Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Jose A L Calbet
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain.,Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain.,Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| |
Collapse
|
33
|
Ensor CM, AlSiraj Y, Shoemaker R, Sturgill J, Keshavamurthy S, Gordon EM, Dong BE, Waters C, Cassis LA. SARS-CoV-2 Spike Protein Regulation of Angiotensin Converting Enzyme 2 and Tissue Renin-Angiotensin Systems: Influence of Biologic Sex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.09.14.460275. [PMID: 34545369 PMCID: PMC8452098 DOI: 10.1101/2021.09.14.460275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Angiotensin converting enzyme 2 (ACE2) is an enzyme that limits activity of the renin-angiotensin system (RAS) and also serves as a receptor for the SARS-CoV-2 Spike (S) protein. Binding of S protein to ACE2 causes internalization which activates local RAS. ACE2 is on the X chromosome and its expression is regulated by sex hormones. In this study, we defined ACE2 mRNA abundance and examined effects of S protein on ACE2 activity and/or angiotensin II (AngII) levels in pivotal tissues (lung, adipose) from male and female mice. In lung, ACE2 mRNA abundance was reduced following gonadectomy (GDX) of male and female mice and was higher in XX than XY mice of the Four Core Genotypes (FCG). Reductions in lung ACE2 mRNA abundance by GDX occurred in XX, but not XY FCG female mice. Lung mRNA abundance of ADAM17 and TMPRSS2, enzymes that shed cell surface ACE2 and facilitate viral cell entry, was reduced by GDX in male but not female mice. For comparison, adipose ACE2 mRNA abundance was higher in female than male mice and higher in XX than XY FCG mice. Adipose ADAM17 mRNA abundance was increased by GDX of male and female mice. S protein reduced ACE2 activity in alveolar type II epithelial cells and 3T3-L1 adipocytes. Administration of S protein to male and female mice increased lung AngII levels and decreased adipose ACE2 activity in male but not female mice. These results demonstrate that sex differences in ACE2 expression levels may impact local RAS following S protein exposures.
Collapse
Affiliation(s)
- Charles M Ensor
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Yasir AlSiraj
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Robin Shoemaker
- Department of Dietetics and Human Nutrition, College of Food, Agriculture and the Environment, University of Kentucky, Lexington, KY 40536
| | - Jamie Sturgill
- Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Suresh Keshavamurthy
- Division of Cardiothoracic Surgery, Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Elizabeth M Gordon
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Brittany E Dong
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Christopher Waters
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
34
|
McClung JA, Levy L, Garcia V, Stec DE, Peterson SJ, Abraham NG. Heme-oxygenase and lipid mediators in obesity and associated cardiometabolic diseases: Therapeutic implications. Pharmacol Ther 2021; 231:107975. [PMID: 34499923 DOI: 10.1016/j.pharmthera.2021.107975] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity-mediated metabolic syndrome remains the leading cause of death worldwide. Among many potential targets for pharmacological intervention, a promising strategy involves the heme oxygenase (HO) system, specifically its inducible form, HO-1. This review collects and updates much of the current knowledge relevant to pharmacology and clinical medicine concerning HO-1 in metabolic diseases and its effect on lipid metabolism. HO-1 has pleotropic effects that collectively reduce inflammation, while increasing vasodilation and insulin and leptin sensitivity. Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). Similarly, HO-1and bilirubin are potential therapeutic targets in the treatment of fat-induced liver diseases. HO-1-mediated upregulation of EET is capable not only of reversing endothelial dysfunction and hypertension, but also of reversing cardiac remodeling, a hallmark of the metabolic syndrome. This process involves browning of white fat tissue (i.e. formation of healthy adipocytes) and reduced lipotoxicity, which otherwise will be toxic to the heart. More importantly, this review examines the activity of EET in biological systems and a series of pathways that explain its mechanism of action and discusses how these might be exploited for potential therapeutic use. We also discuss the link between cardiac ectopic fat deposition and cardiac function in humans, which is similar to that described in obese mice and is regulated by HO-1-EET-PGC1α signaling, a potent negative regulator of the inflammatory adipokine NOV.
Collapse
Affiliation(s)
- John A McClung
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| | - Lior Levy
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States of America
| | - David E Stec
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, United States of America.
| | - Stephen J Peterson
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, United States of America; New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, United States of America
| | - Nader G Abraham
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America; Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States of America.
| |
Collapse
|
35
|
Getachew B, Tizabi Y. Vitamin D and COVID-19: Role of ACE2, age, gender, and ethnicity. J Med Virol 2021; 93:5285-5294. [PMID: 33990955 PMCID: PMC8242434 DOI: 10.1002/jmv.27075] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, disproportionally targets older people, particularly men, ethnic minorities, and individuals with underlying diseases such as compromised immune system, cardiovascular disease, and diabetes. The discrepancy in COVID-19 incidence and severity is multifaceted and likely involves biological, social, as well as nutritional status. Vitamin D deficiency, notably common in Black and Brown people and elderly, is associated with an increased susceptibility to many of the diseases comorbid with COVID-19. Vitamin D deficiency can cause over-activation of the pulmonary renin-angiotensin system (RAS) leading to the respiratory syndrome. RAS is regulated in part at least by angiotensin-converting enzyme 2 (ACE2), which also acts as a primary receptor for SARS-CoV-2 entry into the cells. Hence, vitamin D deficiency can exacerbate COVID-19, via its effects on ACE2. In this review we focus on influence of age, gender, and ethnicity on vitamin D-ACE2 interaction and susceptibility to COVID-19.
Collapse
Affiliation(s)
- Bruk Getachew
- Department of PharmacologyCollege of Medicine, Howard UniversityWashington DCUSA
| | - Yousef Tizabi
- Department of PharmacologyCollege of Medicine, Howard UniversityWashington DCUSA
| |
Collapse
|
36
|
Yanes Cardozo LL, Romero DG. Novel biomarkers of childhood and adolescent obesity. Hypertens Res 2021; 44:1030-1033. [PMID: 33850307 PMCID: PMC8043837 DOI: 10.1038/s41440-021-00651-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 02/28/2021] [Indexed: 02/03/2023]
Abstract
Regulation of the Renin-Angiotensin System (RAS) and the kallikrein-kinin system (KKS) by obesity in adolescents.
Collapse
Affiliation(s)
- Licy L Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA.
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA.
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS, USA.
- Women's Health Research Center, University of Mississippi Medical Center, Jackson, MS, USA.
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Damian G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA.
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS, USA.
- Women's Health Research Center, University of Mississippi Medical Center, Jackson, MS, USA.
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
37
|
Gómez-Zorita S, Milton-Laskibar I, García-Arellano L, González M, Portillo MP. An Overview of Adipose Tissue ACE2 Modulation by Diet and Obesity. Potential Implications in COVID-19 Infection and Severity. Int J Mol Sci 2021; 22:7975. [PMID: 34360741 PMCID: PMC8347022 DOI: 10.3390/ijms22157975] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/18/2021] [Accepted: 07/21/2021] [Indexed: 12/11/2022] Open
Abstract
The present review is aimed at analysing the current evidence concerning the potential modulation of obesity and/or diet in adipose tissue ACE2. Additionally, the potential implications of these effects on COVID-19 are also addressed. The results published show that diet and obesity are two factors that effectively influence the expression of Ace2 gene in adipose tissue. However, the shifts in this gene do not always occur in the same direction, nor with the same intensity. Additionally, there is no consensus regarding the implications of increased adipose tissue ACE2 expression in health. Thus, while in some studies a protective role is attributed to ACE2 overexpression, other studies suggest otherwise. Similarly, there is much debate regarding the role played by ACE2 in COVID-19 in terms of degree of infection and disease outcomes. The greater risk of infection that may hypothetically derive from enhanced ACE2 expression is not clear since the functionality of the enzyme seems to be as important as the abundance. Thus, the greater abundance of ACE2 in adipose tissue of obese subjects may be counterbalanced by its lower activation. In addition, a protective role of ACE2 overexpression has also been suggested, associated with the increase in anti-inflammatory factors that it may produce.
Collapse
Affiliation(s)
- Saioa Gómez-Zorita
- Nutrition and Obesity Group, Lucio Lascaray Research Center, Department of Nutrition and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain; (L.G.-A.); (M.P.P.)
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- BIOARABA Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Iñaki Milton-Laskibar
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, 28049 Madrid, Spain
| | - Laura García-Arellano
- Nutrition and Obesity Group, Lucio Lascaray Research Center, Department of Nutrition and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain; (L.G.-A.); (M.P.P.)
| | - Marcela González
- Nutrition and Food Science Department, Faculty of Biochemistry and Biological Sciences, National University of Litoral and National Scientific and Technical Research Council (CONICET), Santa Fe 3000, Argentina;
| | - María P. Portillo
- Nutrition and Obesity Group, Lucio Lascaray Research Center, Department of Nutrition and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain; (L.G.-A.); (M.P.P.)
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- BIOARABA Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| |
Collapse
|
38
|
Bastolla U. Mathematical Model of SARS-Cov-2 Propagation Versus ACE2 Fits COVID-19 Lethality Across Age and Sex and Predicts That of SARS. Front Mol Biosci 2021; 8:706122. [PMID: 34322518 PMCID: PMC8311794 DOI: 10.3389/fmolb.2021.706122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
The fatality rate of Covid-19 escalates with age and is larger in men than women. I show that these variations correlate strongly with the level of the viral receptor protein ACE2 in rat lungs, which is consistent with the still limited data on human ACE2. Surprisingly, lower receptor levels correlate with higher fatality. I propose two possible explanations of this negative correlation: First, a previous mathematical model predicts that the velocity of viral progression in the organism as a function of the receptor level has a maximum and declines for abundant receptor. Secondly, degradation of ACE2 by the virus may cause the runaway inflammatory response that characterizes severe CoViD-19. I present here a mathematical model that predicts the lethality as a function of ACE2 protein level based on the two above hypothesis. The model fits Covid-19 fatality rate across age and sex in three countries with high accuracy (r 2 > 0.9 ) under the hypothesis that the speed of viral progression in the infected organism is a decreasing function of the ACE2 level. Moreover, rescaling the fitted parameters by the ratio of the binding rates of the spike proteins of SARS-CoV and SARS-CoV-2 allows predicting the fatality rate of SARS-CoV across age and sex, thus linking the molecular and epidemiological levels.
Collapse
Affiliation(s)
- Ugo Bastolla
- Centro de Biologia Molecular “Severo Ochoa”, CSIC-UAM Cantoblanco, Madrid, Spain
| |
Collapse
|
39
|
Liu K, Chen X, Ren X, Wu Y, Ren S, Qin C. SARS-CoV-2 effects in the genitourinary system and prospects of sex hormone therapy. Asian J Urol 2021; 8:303-314. [PMID: 33282690 PMCID: PMC7703223 DOI: 10.1016/j.ajur.2020.11.004] [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: 06/16/2020] [Accepted: 08/20/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECT Corona virus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which leads to acute respiratory infection symptoms. SARS-CoV-2 infection is not always limited to the respiratory tract, and renal infection and dysfunction have been shown to be specific risk factors for death. In addition, COVID-19 has a higher incidence, severity and mortality in men than women. This disparity is due to biological rather than comorbid or behavioral sex differences. Because the male reproductive system is unique, the function of sex hormones in COVID-19 infection may explain the differences between males and females. Understanding these factors will provide appropriate prevention measures and adequate triage strategies and guide the drug discovery process. METHODS An electronic search was completed in PubMed, ARXIV, MEDRXIV and BIORXIV. The most relevant articles were systematically reviewed. In addition, single cell RNA sequencing analysis of tissue samples from human cell landscape was conducted. RESULTS The influence of SARS-CoV-2 on the urogenital system, the possibility of urinary tract transmission and the functions of sex hormones were discussed in this review. CONCLUSION Corona viruses can invade the genitourinary system, causing urological symptoms. Identifying the potential genitourinary organ impairments and protecting them from damage are necessary. Since sex hormones have potential as specific drugs, the gonadal hormones substitution therapy should be considered in both sexes in the COVID-19 pandemic.
Collapse
Affiliation(s)
| | - Xinglin Chen
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaohan Ren
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuqing Wu
- Medical College, Southeast University, Nanjing, China
| | - Shancheng Ren
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chao Qin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Mehay D, Silberman Y, Arnold AC. The Arcuate Nucleus of the Hypothalamus and Metabolic Regulation: An Emerging Role for Renin-Angiotensin Pathways. Int J Mol Sci 2021; 22:7050. [PMID: 34208939 PMCID: PMC8268643 DOI: 10.3390/ijms22137050] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 12/02/2022] Open
Abstract
Obesity is a chronic state of energy imbalance that represents a major public health problem and greatly increases the risk for developing hypertension, hyperglycemia, and a multitude of related pathologies that encompass the metabolic syndrome. The underlying mechanisms and optimal treatment strategies for obesity, however, are still not fully understood. The control of energy balance involves the actions of circulating hormones on a widely distributed network of brain regions involved in the regulation of food intake and energy expenditure, including the arcuate nucleus of the hypothalamus. While obesity is known to disrupt neurocircuits controlling energy balance, including those in the hypothalamic arcuate nucleus, the pharmacological targeting of these central mechanisms often produces adverse cardiovascular and other off-target effects. This highlights the critical need to identify new anti-obesity drugs that can activate central neurocircuits to induce weight loss without negatively impacting blood pressure control. The renin-angiotensin system may provide this ideal target, as recent studies show this hormonal system can engage neurocircuits originating in the arcuate nucleus to improve energy balance without elevating blood pressure in animal models. This review will summarize the current knowledge of renin-angiotensin system actions within the arcuate nucleus for control of energy balance, with a focus on emerging roles for angiotensin II, prorenin, and angiotensin-(1-7) pathways.
Collapse
Affiliation(s)
| | | | - Amy C. Arnold
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.M.); (Y.S.)
| |
Collapse
|
41
|
Obesity-associated cardiovascular risk in women: hypertension and heart failure. Clin Sci (Lond) 2021; 135:1523-1544. [PMID: 34160010 DOI: 10.1042/cs20210384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/14/2021] [Accepted: 06/07/2021] [Indexed: 02/07/2023]
Abstract
The pathogenesis of obesity-associated cardiovascular diseases begins long prior to the presentation of a cardiovascular event. In both men and women, cardiovascular events, and their associated hospitalizations and mortality, are often clinically predisposed by the presentation of a chronic cardiovascular risk factor. Obesity increases the risk of cardiovascular diseases in both sexes, however, the clinical prevalence of obesity, as well as its contribution to crucial cardiovascular risk factors is dependent on sex. The mechanisms via which obesity leads to cardiovascular risk is also discrepant in women between their premenopausal, pregnancy and postmenopausal phases of life. Emerging data indicate that at all reproductive statuses and ages, the presentation of a cardiovascular event in obese women is strongly associated with hypertension and its subsequent chronic risk factor, heart failure with preserved ejection fraction (HFpEF). In addition, emerging evidence indicates that obesity increases the risk of both hypertension and heart failure in pregnancy. This review will summarize clinical and experimental data on the female-specific prevalence and mechanisms of hypertension and heart failure in women across reproductive stages and highlight the particular risks in pregnancy as well as emerging data in a high-risk ethnicity in women of African ancestry (AA).
Collapse
|
42
|
Terada M, Ohtsu H, Saito S, Hayakawa K, Tsuzuki S, Asai Y, Matsunaga N, Kutsuna S, Sugiura W, Ohmagari N. Risk factors for severity on admission and the disease progression during hospitalisation in a large cohort of patients with COVID-19 in Japan. BMJ Open 2021; 11:e047007. [PMID: 34130961 PMCID: PMC8210659 DOI: 10.1136/bmjopen-2020-047007] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES To investigate the risk factors contributing to severity on admission. Additionally, risk factors of worst severity and fatality were studied. Moreover, factors were compared based on three points: early severity, worst severity and fatality. DESIGN An observational cohort study using data entered in a Japan nationwide COVID-19 inpatient registry, COVIREGI-JP. SETTING As of 28 September 2020, 10480 cases from 802 facilities have been registered. Participating facilities cover a wide range of hospitals where patients with COVID-19 are admitted in Japan. PARTICIPANTS Participants who had a positive test result on any applicable SARS-CoV-2 diagnostic tests were admitted to participating healthcare facilities. A total of 3829 cases were identified from 16 January to 31 May 2020, of which 3376 cases were included in this study. PRIMARY AND SECONDARY OUTCOME MEASURES Primary outcome was severe or nonsevere on admission, determined by the requirement of mechanical ventilation or oxygen therapy, SpO2 or respiratory rate. Secondary outcome was the worst severity during hospitalisation, judged by the requirement of oxygen and/orinvasive mechanical ventilation/extracorporeal membrane oxygenation. RESULTS Risk factors for severity on admission were older age, men, cardiovascular disease, chronic respiratory disease, diabetes, obesity and hypertension. Cerebrovascular disease, liver disease, renal disease or dialysis, solid tumour and hyperlipidaemia did not influence severity on admission; however, it influenced worst severity. Fatality rates for obesity, hypertension and hyperlipidaemia were relatively lower. CONCLUSIONS This study segregated the comorbidities influencing severity and death. It is possible that risk factors for severity on admission, worst severity and fatality are not consistent and may be propelled by different factors. Specifically, while hypertension, hyperlipidaemia and obesity had major effect on worst severity, their impact was mild on fatality in the Japanese population. Some studies contradict our results; therefore, detailed analyses, considering in-hospital treatments, are needed for validation. TRIAL REGISTRATION NUMBER UMIN000039873. https://upload.umin.ac.jp/cgi-open-bin/ctr_e/ctr_view.cgi?recptno=R000045453.
Collapse
Affiliation(s)
- Mari Terada
- Department of Infectious Diseases, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroshi Ohtsu
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Sho Saito
- Department of Infectious Diseases, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kayoko Hayakawa
- Department of Infectious Diseases, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinya Tsuzuki
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Yusuke Asai
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Nobuaki Matsunaga
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Satoshi Kutsuna
- Department of Infectious Diseases, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Wataru Sugiura
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Department of Infectious Diseases, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
43
|
Miličić Stanić B, Maddox S, de Souza AMA, Wu X, Mehranfard D, Ji H, Speth RC, Sandberg K. Male bias in ACE2 basic science research: missed opportunity for discovery in the time of COVID-19. Am J Physiol Regul Integr Comp Physiol 2021; 320:R925-R937. [PMID: 33848207 PMCID: PMC8203415 DOI: 10.1152/ajpregu.00356.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 11/22/2022]
Abstract
Throughout the world, including the United States, men have worse outcomes from COVID-19 than women. SARS-CoV-2, the causative virus of the COVID-19 pandemic, uses angiotensin-converting enzyme 2 (ACE2) to gain cellular entry. ACE2 is a member of the renin-angiotensin system (RAS) and plays an important role in counteracting the harmful effects mediated by the angiotensin type 1 receptor. Therefore, we conducted Ovid MEDLINE and Embase database searches of basic science studies investigating the impact of the biological variable of sex on ACE2 expression and regulation from 2000, the year ACE2 was discovered, through December 31, 2020. Out of 2,131 publications, we identified 853 original research articles on ACE2 conducted in primary cells, tissues, and/or whole mammals excluding humans. The majority (68.7%) of these studies that cited the sex of the animal were conducted in males, while 11.2% were conducted solely in females; 9.26% compared ACE2 between the sexes, while 10.8% did not report the sex of the animals used. General findings are that sex differences are tissue-specific and when present, are dependent upon gonadal state. Renal, cardiac, and adipose ACE2 is increased in both sexes under experimental conditions that model co-morbidities associated with worse COVID-19 outcomes including hypertension, obesity, and renal and cardiovascular diseases; however, ACE2 protein was generally higher in the males. Studies in Ace2 knockout mice indicate ACE2 plays a greater role in protecting the female from developing hypertension than the male. Studying the biological variable of sex in ACE2 research provides an opportunity for discovery in conditions involving RAS dysfunction and will shed light on sex differences in COVID-19 severity.
Collapse
Affiliation(s)
- Branka Miličić Stanić
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Sydney Maddox
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Aline M A de Souza
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Xie Wu
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Danial Mehranfard
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida
| | - Hong Ji
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Robert C Speth
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, District of Columbia
| | - Kathryn Sandberg
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| |
Collapse
|
44
|
Sugawara A, Shimada H, Otsubo Y, Kouketsu T, Suzuki S, Yokoyama A. The usefulness of angiotensin-(1-7) and des-Arg 9-bradykinin as novel biomarkers for metabolic syndrome. Hypertens Res 2021; 44:1034-1036. [PMID: 34045691 DOI: 10.1038/s41440-021-00671-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Hiroki Shimada
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yuri Otsubo
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takumi Kouketsu
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Susumu Suzuki
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Yokoyama
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
45
|
Zhang J, Huang F, Xia B, Yuan Y, Yu F, Wang G, Chen Q, Wang Q, Li Y, Li R, Song Z, Pan T, Chen J, Lu G, Zhang H. The interferon-stimulated exosomal hACE2 potently inhibits SARS-CoV-2 replication through competitively blocking the virus entry. Signal Transduct Target Ther 2021; 6:189. [PMID: 33980808 PMCID: PMC8113286 DOI: 10.1038/s41392-021-00604-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/25/2021] [Accepted: 03/21/2021] [Indexed: 01/08/2023] Open
Abstract
Since the outbreak of coronavirus disease 2019 (COVID-19), it has become a global pandemic. The spike (S) protein of etiologic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) specifically recognizes human angiotensin-converting enzyme 2 (hACE2) as its receptor, which is recently identified as an interferon (IFN)-stimulated gene. Here, we find that hACE2 exists on the surface of exosomes released by different cell types, and the expression of exosomal hACE2 is increased by IFNα/β treatment. In particular, exosomal hACE2 can specifically block the cell entry of SARS-CoV-2, subsequently inhibit the replication of SARS-CoV-2 in vitro and ex vivo. Our findings have indicated that IFN is able to upregulate a viral receptor on the exosomes which competitively block the virus entry, exhibiting a potential antiviral strategy.
Collapse
Affiliation(s)
- Junsong Zhang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Feng Huang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
| | - Baijin Xia
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yaochang Yuan
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fei Yu
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guanwen Wang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Qianyu Chen
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Qian Wang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yuzhuang Li
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Rong Li
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zheng Song
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ting Pan
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jingliang Chen
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Infectious Disease Center, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Gen Lu
- Department of Respiratory Diseases, Guangzhou Women and Children Hospital, Guangzhou, Guangdong, China
| | - Hui Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
46
|
Flinn B, Royce N, Gress T, Chowdhury N, Santanam N. Dual role for angiotensin-converting enzyme 2 in Severe Acute Respiratory Syndrome Coronavirus 2 infection and cardiac fat. Obes Rev 2021; 22:e13225. [PMID: 33660398 PMCID: PMC8013367 DOI: 10.1111/obr.13225] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/25/2021] [Accepted: 02/04/2021] [Indexed: 12/15/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been an increasingly prevalent target for investigation since its discovery 20 years ago. The finding that it serves a counterregulatory function within the traditional renin-angiotensin system, implicating it in cardiometabolic health, has increased its clinical relevance. Focus on ACE2's role in cardiometabolic health has largely centered on its apparent functions in the context of obesity. Interest in ACE2 has become even greater with the discovery that it serves as the cell receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), opening up numerous mechanisms for deleterious effects of infection. The proliferation of ACE2 within the literature coupled with its dual role in SARS-CoV-2 infection and obesity necessitates review of the current understanding of ACE2's physiological, pathophysiological, and potential therapeutic functions. This review highlights the roles of ACE2 in cardiac dysfunction and obesity, with focus on epicardial adipose tissue, to reconcile the data in the context of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Brendin Flinn
- Department of Biomedical Sciences, Joan C Edwards School of Medicine, Huntington, West Virginia, USA
| | - Nicholas Royce
- Department of Biomedical Sciences, Joan C Edwards School of Medicine, Huntington, West Virginia, USA
| | - Todd Gress
- Research Service, Hershel "Woody" Williams VA Medical Center, Huntington, West Virginia, USA
| | - Nepal Chowdhury
- Department of Cardiovascular and Thoracic Surgery, St. Mary's Heart Center, Huntington, WV, USA
| | - Nalini Santanam
- Department of Biomedical Sciences, Joan C Edwards School of Medicine, Huntington, West Virginia, USA
| |
Collapse
|
47
|
Cao P, Song Y, Zhuang Z, Ran J, Xu L, Geng Y, Han L, Zhao S, Qin J, He D, Wu F, Yang L. Obesity and COVID-19 in Adult Patients With Diabetes. Diabetes 2021; 70:1061-1069. [PMID: 33597204 DOI: 10.2337/db20-0671] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 02/12/2021] [Indexed: 01/08/2023]
Abstract
Obesity has caused wide concerns due to its high prevalence in patients with severe coronavirus disease 2019 (COVID-19). Coexistence of diabetes and obesity could cause an even higher risk of severe outcomes due to immunity dysfunction. We conducted a retrospective study in 1,637 adult patients who were admitted into an acute hospital in Wuhan, China. Propensity score-matched logistic regression was used to estimate the risks of severe pneumonia and requiring in-hospital oxygen therapy associated with obesity. After adjustment for age, sex, and comorbidities, obesity was significantly associated with higher odds of severe pneumonia (odds ratio [OR] 1.47 [95% CI 1.15-1.88]; P = 0.002) and oxygen therapy (OR 1.40 [95% CI 1.10-1.79]; P = 0.007). Higher ORs of severe pneumonia due to obesity were observed in men, older adults, and those with diabetes. Among patients with diabetes, overweight increased the odds of requiring in-hospital oxygen therapy by 0.68 times (P = 0.014) and obesity increased the odds by 1.06 times (P = 0.028). A linear dose-response curve between BMI and severe outcomes was observed in all patients, whereas a U-shaped curve was observed in those with diabetes. Our findings provide important evidence to support obesity as an independent risk factor for severe outcomes of COVID-19 infection in the early phase of the ongoing pandemic.
Collapse
Affiliation(s)
- Peihua Cao
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Song
- Department of Medical Affairs, Huoshenshan Hospital, Wuhan, China
- No. 923 Hospital of Joint Service Supporting Force, Nanning, China
| | - Zian Zhuang
- Department of Applied Mathematics, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Jinjun Ran
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Xu
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yan Geng
- Department of Infectious Diseases, Huoshenshan Hospital, Wuhan, China
- Department of Gastroenterology, No. 923 Hospital of Joint Service Supporting Force, Nanning, China
| | - Lefei Han
- School of Nursing, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shi Zhao
- Jockey Club School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jing Qin
- School of Nursing, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Daihai He
- Department of Applied Mathematics, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Fengfu Wu
- Department of Medical Affairs, Huoshenshan Hospital, Wuhan, China
- No. 923 Hospital of Joint Service Supporting Force, Nanning, China
| | - Lin Yang
- School of Nursing, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| |
Collapse
|
48
|
Foresta C, Rocca MS, Di Nisio A. Gender susceptibility to COVID-19: a review of the putative role of sex hormones and X chromosome. J Endocrinol Invest 2021; 44:951-956. [PMID: 32936429 PMCID: PMC7492232 DOI: 10.1007/s40618-020-01383-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/31/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND The recent emergence of COVID-19 poses a global health emergency. One of the most frequently reported data is sex-related severity and mortality: according to the last available analysis on 239,709 patients in Italy, lethality is 17.7% in men and 10.8% in women, with 59% of total deaths being men. Interestingly, the infection rate is lower in males than in females, with 45.8% and 54.2% of positive cases, respectively, suggesting that gender-related factor may worsen disease evolution. A tentative hypothesis to explain these findings is the role of angiotensin-converting enzyme 2 (ACE2) and serine protease TMPRSS2 involved in viral infection. PURPOSE In this review, we summarize the available evidence pointing to gender-related differences in ACE2 and TMPRSS2 expression, from both genetic and endocrine points of view. RESULTS Altogether, available evidence points toward two not-mutually exclusive mechanisms in gender susceptibility to COVID-19 by sex hormonal regulation of ACE2 and TMPRSS2. On one hand, ACE2 expression could be increased in women, either by estrogens or constitutively by X chromosome inactivation escape or by reduced methylation, providing a larger reservoir of ACE2 to maintain the fundamental equilibrium of RAS regulatory axis. On the other, low levels of androgens in women may keep at low levels TMPRSS2 expression, representing a further protective factor for the development of COVID-19 infection, despite the increased expression of ACE2, which represents the Trojan horse for SARS-CoV-2 entry. CONCLUSIONS Both mechanisms consistently point to the role of sex hormones and sex chromosomes in the differential severity and lethality of COVID-19 in men and women.
Collapse
Affiliation(s)
- C Foresta
- Department of Medicine, Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy.
| | - M S Rocca
- Department of Medicine, Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy
| | - A Di Nisio
- Department of Medicine, Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy
| |
Collapse
|
49
|
Aleksova A, Gagno G, Sinagra G, Beltrami AP, Janjusevic M, Ippolito G, Zumla A, Fluca AL, Ferro F. Effects of SARS-CoV-2 on Cardiovascular System: The Dual Role of Angiotensin-Converting Enzyme 2 (ACE2) as the Virus Receptor and Homeostasis Regulator-Review. Int J Mol Sci 2021; 22:4526. [PMID: 33926110 PMCID: PMC8123609 DOI: 10.3390/ijms22094526] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/22/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is the entry receptor for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the cause of Coronavirus Disease-2019 (COVID-19) in humans. ACE-2 is a type I transmembrane metallocarboxypeptidase expressed in vascular endothelial cells, alveolar type 2 lung epithelial cells, renal tubular epithelium, Leydig cells in testes and gastrointestinal tract. ACE2 mediates the interaction between host cells and SARS-CoV-2 spike (S) protein. However, ACE2 is not only a SARS-CoV-2 receptor, but it has also an important homeostatic function regulating renin-angiotensin system (RAS), which is pivotal for both the cardiovascular and immune systems. Therefore, ACE2 is the key link between SARS-CoV-2 infection, cardiovascular diseases (CVDs) and immune response. Susceptibility to SARS-CoV-2 seems to be tightly associated with ACE2 availability, which in turn is determined by genetics, age, gender and comorbidities. Severe COVID-19 is due to an uncontrolled and excessive immune response, which leads to acute respiratory distress syndrome (ARDS) and multi-organ failure. In spite of a lower ACE2 expression on cells surface, patients with CVDs have a higher COVID-19 mortality rate, which is likely driven by the imbalance between ADAM metallopeptidase domain 17 (ADAM17) protein (which is required for cleavage of ACE-2 ectodomain resulting in increased ACE2 shedding), and TMPRSS2 (which is required for spike glycoprotein priming). To date, ACE inhibitors and Angiotensin II Receptor Blockers (ARBs) treatment interruption in patients with chronic comorbidities appears unjustified. The rollout of COVID-19 vaccines provides opportunities to study the effects of different COVID-19 vaccines on ACE2 in patients on treatment with ACEi/ARB.
Collapse
Affiliation(s)
- Aneta Aleksova
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (G.G.); (G.S.); (M.J.); (A.L.F.); (F.F.)
| | - Giulia Gagno
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (G.G.); (G.S.); (M.J.); (A.L.F.); (F.F.)
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (G.G.); (G.S.); (M.J.); (A.L.F.); (F.F.)
| | | | - Milijana Janjusevic
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (G.G.); (G.S.); (M.J.); (A.L.F.); (F.F.)
| | - Giuseppe Ippolito
- National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00135 Rome, Italy;
| | - Alimuddin Zumla
- Department of Infection, Division of Infection and Immunity, Centre for Clinical Microbiology, University College London, London NW3 2PF, UK;
- National Institute for Health Research Biomedical Research Centre, University College London Hospitals, London NW1 2BU, UK
| | - Alessandra Lucia Fluca
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (G.G.); (G.S.); (M.J.); (A.L.F.); (F.F.)
| | - Federico Ferro
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (G.G.); (G.S.); (M.J.); (A.L.F.); (F.F.)
| |
Collapse
|
50
|
Huang A, Kandhi S, Sun D. Roles of Genetic Predisposition in the Sex Bias of Pulmonary Pathophysiology, as a Function of Estrogens : Sex Matters in the Prevalence of Lung Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:107-127. [PMID: 33788190 DOI: 10.1007/978-3-030-63046-1_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In addition to studies focused on estrogen mediation of sex-different regulation of systemic circulations, there is now increasing clinical relevance and research interests in the pulmonary circulation, in terms of sex differences in the morbidity and mortality of lung diseases such as inherent-, allergic- and inflammatory-based events. Thus, female predisposition to pulmonary artery hypertension (PAH) is an inevitable topic. To better understand the nature of sexual differentiation in the pulmonary circulation, and how heritable factors, in vivo- and/or in vitro-altered estrogen circumstances and changes in the live environment work in concert to discern the sex bias, this chapter reviews pulmonary events characterized by sex-different features, concomitant with exploration of how alterations of genetic expression and estrogen metabolisms trigger the female-predominant pathological signaling. We address the following: PAH (Sect.7.2) is characterized as an estrogenic promotion of its incidence (Sect. 7.2.2), as a function of specific germline mutations, and as an estrogen-elicited protection of its prognosis (Sect.7.2.1). More detail is provided to introduce a less recognized gene of Ephx2 that encodes soluble epoxide hydrolase (sEH) to degrade epoxyeicosatrienic acids (EETs). As a susceptible target of estrogen, Ephx2/sEH expression is downregulated by an estrogen-dependent epigenetic mechanism. Increases in pulmonary EETs then evoke a potentiation of PAH generation, but mitigation of its progression, a phenomenon similar to the estrogen-paradox regulation of PAH. Additionally, the female susceptibility to chronic obstructive pulmonary diseases (Sect. 7.3) and asthma (Sect.7.4), but less preference to COVID-19 (Sect. 7.5), and roles of estrogen in their pathogeneses are briefly discussed.
Collapse
Affiliation(s)
- An Huang
- Department of Physiology, New York Medical College, Valhalla, NY, USA.
| | - Sharath Kandhi
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|