1
|
Takeuchi LE, Kalia LV, Simmons CA. Vascular models of Alzheimer's disease: An overview of recent in vitro models of the blood-brain barrier. Neurobiol Dis 2025; 208:106864. [PMID: 40089165 DOI: 10.1016/j.nbd.2025.106864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) remains an overwhelming epidemiologic and economic burden on our healthcare systems, affecting an estimate of 11 % of individuals aged 65 years and older. Increasing evidence of the role of the blood-brain barrier (BBB) in AD pathology lends support to the vascular hypothesis of AD, which posits that damage to cerebral vasculature and impairments to cerebral blood flow are major contributors to neurodegeneration in AD. While the question remains whether the dysfunction of the BBB is the cause or consequence of the disease, understanding of the relationship between vascular pathology and AD is growing increasingly complex, warranting the need for better tools to study vasculature in AD. This review provides an overview of AD models in the context of studying vascular impairments and their relevance in pathology. Specifically, we summarize opportunities in in vitro models, cell sources, and phenotypic observations in sporadic and familial forms of AD. Further, we describe recent advances in generating models which recapitulate in vivo characteristics of the BBB in AD through the use of microfluidics, induced pluripotent stem cells (iPSC), and organoid technologies. Finally, we provide a searchable database of reported cell-based models of pathogenic AD gene variants.
Collapse
Affiliation(s)
- Lily E Takeuchi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Lorraine V Kalia
- Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada; Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| |
Collapse
|
2
|
Xu Z, Yu H, Zhuang R, Fan Q. Immunotherapy for hypertensive end-organ damage: a new therapeutic strategy. Essays Biochem 2025; 0:EBC20243000. [PMID: 40134277 DOI: 10.1042/ebc20243000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/07/2025] [Indexed: 03/27/2025]
Abstract
Hypertension represents a highly prevalent chronic condition and stands among the foremost contributors to premature mortality on a global scale. Its etiopathogenesis is intricate and multifaceted, being shaped by a diverse array of elements such as age, genetic predisposition, and activation of the neuroendocrine apparatus. Mounting evidence has shed light on the significant part that autoimmune responses play in hypertension and the ensuing damage to end organs. Virtually all varieties of immune cells, spanning both innate and adaptive immune compartments, exhibit a close correlation with the progression of hypertension. These immune cells infiltrate the kidney and vascular mesenchyme, subsequently discharging potent cytokines, reactive oxygen species, and metalloproteinases. This cascade of events can affect the functionality of local blood vessels and potentially precipitate adverse structural and functional alterations in crucial organs like the heart and kidney. In recent times, the management of end-organ damage in hypertension has emerged as a pivotal scientific focus. A multitude of researchers are actively engaged in probing efficacious intervention regimens, among which immunotherapy strategies hold considerable promise and anticipation as a prospective avenue.
Collapse
Affiliation(s)
- Zhiyang Xu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210000, China
- Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Haisheng Yu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210000, China
| | - Rulin Zhuang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Qin Fan
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210000, China
| |
Collapse
|
3
|
Wiley CR, Williams DP, Sigrist C, Brownlow BN, Markser A, Hong S, Sternberg EM, Kapuku G, Koenig J, Thayer JF. Differences in inflammation among black and white individuals: A systematic review and meta-analysis. Brain Behav Immun 2025; 127:269-286. [PMID: 40101808 DOI: 10.1016/j.bbi.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 02/13/2025] [Accepted: 03/09/2025] [Indexed: 03/20/2025] Open
Abstract
IMPORTANCE Despite persisting health disparities between Black and White individuals, racial differences in inflammation have yet to be comprehensively examined. OBJECTIVE To determine if significant differences in circulating levels of inflammatory markers between Black and White populations exist. DATA SOURCES Studies were identified through systematic searches of four electronic databases in January 2022. Additional studies were identified via reference lists and e-mail contact. STUDY SELECTION Eligible studies included full-text empirical articles that consisted of Black and White individuals and reported statistics for inflammatory markers for each racial group. Of the 1368 potentially eligible studies, 84 (6.6 %) representing more than one million participants met study selection criteria. DATA EXTRACTION AND SYNTHESIS Risk of bias was assessed via meta regressions that considered relevant covariates. Data heterogeneity was tested using both the Cochrane Q-statistic and the standard I2 index. Random effects models were used to calculate estimates of effect size from standardized mean differences. MAIN OUTCOMES AND MEASURES Outcome measures included 12 inflammatory markers, including C-reactive protein (CRP), Fibrinogen, Interleukin-6 (IL-6), Tumor necrosis factor-alpha (TNF-α), and soluble intercellular adhesion molecule 1 (sICAM-1). RESULTS Several markers had robust sample sizes for analysis, including CRP (White N = 934,594; Black N = 55,234), Fibrinogen (White N = 80,880; Black N = 18,001), and IL-6 (White N = 20,269; Black N = 14,675). Initial results indicated significant effects on CRP (k = 56, pooled Hedges' g = 0.24), IL-6 (k = 33, g = 0.15), and Fibrinogen (k = 19, g = 0.49), with Black individuals showing higher levels. Results also indicated significant effects on sICAM-1 (k = 6, g = -0.46), and Interleukin-10 (k = 4, g = -0.18), with White individuals showing higher levels. Sensitivity analyses confirmed robust effects for CRP, IL-6, Fibrinogen, and sICAM-1 while also revealing significant effects on TNF-α (k = 18, g = -0.17) and Interleukin-8 (k = 5, g = -0.19), with White individuals showing higher levels of both. CONCLUSIONS AND RELEVANCE Current meta-analytic results provide evidence for marked racial differences in common circulating inflammatory markers and illustrate the complexity of the inflammatory profile differences between Black and White individuals. Review Pre-Registration: PROSPERO Identifier - CRD42022312352.
Collapse
Affiliation(s)
- Cameron R Wiley
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Psychological Science, University of California, Irvine, Irvine, CA, United States.
| | - DeWayne P Williams
- Department of Psychological Science, University of California, Irvine, Irvine, CA, United States
| | - Christine Sigrist
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, United States
| | - Briana N Brownlow
- Duke University Medical Center, Duke University, Durham, NC, United States
| | - Anna Markser
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, United States
| | - Suzi Hong
- Herbert Wertheim School of Public Health, University of California, San Diego, San Diego, CA, United States; Department of Psychiatry, School of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Esther M Sternberg
- Center for Integrative Medicine, The University of Arizona, Tucson, AZ, United States
| | - Gaston Kapuku
- Department of Pediatrics and Medicine, Georgia Prevention Institute, Augusta, GA, United States
| | - Julian Koenig
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, United States
| | - Julian F Thayer
- Department of Psychological Science, University of California, Irvine, Irvine, CA, United States; Department of Psychology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
4
|
Tang Y, Shi J, Wang L, Zhang Y, Xu L, Sun T. Development of a predictive model for immune‑related adverse events in patients with cancer. Oncol Lett 2025; 29:103. [PMID: 39736926 PMCID: PMC11683521 DOI: 10.3892/ol.2024.14849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
It is crucial to accurately identify patients with cancer at high risk for immune-related adverse events (irAEs) caused by immune checkpoint inhibitors (ICIs). The present retrospective study analyzed the risk factors for irAEs in 992 patients with cancer treated with ICIs at Xi'an International Medical Center Hospital from December 2021 to December 2023. The patients were categorized into one group that experienced irAEs (n=276) and a control group (n=716) based on the occurrence of irAEs. The clinical characteristics of irAEs group (n=276) and control group (n=716) were analyzed to identify the risk factors of irAEs in patients with cancer. Multivariate regression analysis revealed significant differences between the two groups in terms of hypertension, primary cancer, metastasis, targeted drug combination and radiotherapy (P<0.05). A nomogram predictive model for irAEs was developed based on the relevant risk factors. The predictive model for irAEs in patients with cancer yielded an area under the receiver operating characteristic (ROC) curve of 0.672 (95% confidence interval: 0.630-0.714). In the validation set, the Hosmer-Lemeshow goodness-of-fit test demonstrated a favorable fit with a chi-square value of 0.787 and a P-value of 0.978. The developed predictive model can effectively identify high-risk patients with irAEs, facilitate early identification of irAEs, thereby optimizing the management strategies of irAEs, and ultimately improving the quality of life for patients.
Collapse
Affiliation(s)
- Yajuan Tang
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Jinping Shi
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Liping Wang
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Yan Zhang
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Liting Xu
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Tao Sun
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
5
|
Watanabe DK, Kitayama S, Williams DP, Thayer JF. Emotion suppression differentially moderates the link between stress and cardiovascular disease risk in Japanese and Americans. Int J Clin Health Psychol 2025; 25:100555. [PMID: 40110442 PMCID: PMC11919598 DOI: 10.1016/j.ijchp.2025.100555] [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: 12/11/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Background Cardiovascular disease (CVD) remains a key cause of mortality worldwide. Prior work has found that the association between stress and cardiovascular outcomes is moderated by emotion regulation (ER) and expressive suppression (i.e., emotion inhibition), which is linked with adverse outcomes (i.e., inflammation) in Western (Americans) but not Eastern (Japanese) populations. Existing cultural differences in biological stress responses and suppression use suggest that these factors may have different implications for CV outcomes. Objective We address this gap in the literature by examining if ER differentially moderates the relationship between stress and CVD risk between Japanese and American adults. Method Participants were from the Midlife in Japan and Midlife in the United States studies and had complete biomarker and psychological data (Japanese: N = 315, M age = 59.22, 149 females; Americans: N = 524, M age = 51.98, 291 females). Stress was indexed using the perceived stress scale. Trait suppression and reappraisal were indexed using the Emotion Regulation Questionnaire. CVD risk was indexed using a composite score of body mass index, C-reactive protein, interleukin-6, systolic blood pressure, and high-density lipoprotein cholesterol ratio. Results Adjusting for age, sex, education, tobacco, alcohol, and prescription medication use, linear regressions revealed robust cultural differences among those with high suppression (r = -0.10 [-0.19, -0.01]). Higher stress was linked with higher CVD risk in Americans regardless of the level of reappraisal or suppression (r's > 0.11, p's < 0.07). In contrast, among Japanese with high suppression, higher stress was associated with lower CVD risk (r = -0.09 [-0.23, 0.05]). Higher stress was associated with greater inflammation among Japanese with lower suppression (r = 0.10 [-0.07, 0.28]). Conclusions Consistent with prior work, these findings suggest that adaptive ER moderates the association between stress and CVD risk, and that suppression may not be universally 'maladaptive.' Results emphasize the importance of considering cultural context when assessing the impact of emotion suppression on health, which may help explain differences in CVD outcomes between individuals from Eastern and Western populations.
Collapse
Affiliation(s)
- Darcianne K Watanabe
- School of Social Ecology, c/o Department of Psychological Science, University of California, Irvine, 4201 Social and Behavioral Sciences Gateway, Irvine, CA, 92697, USA
| | - Shinobu Kitayama
- Department of Psychology, University of Michigan, 1004 East Hall, 530 Church Street, Ann Arbor, MI, 48109, USA
| | - DeWayne P Williams
- Department of Psychological Science, University of California, Irvine, 4201 Social and Behavioral Sciences Gateway, Irvine, CA, 92697, USA
| | - Julian F Thayer
- Department of Psychological Science, University of California, Irvine, 4201 Social and Behavioral Sciences Gateway, Irvine, CA, 92697, USA
| |
Collapse
|
6
|
Goorani S, Zangene S, Imig JD. Hypertension: A Continuing Public Healthcare Issue. Int J Mol Sci 2024; 26:123. [PMID: 39795981 PMCID: PMC11720251 DOI: 10.3390/ijms26010123] [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: 11/06/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Hypertension is a cardiovascular disease defined by an elevated systemic blood pressure. This devastating disease afflicts 30-40% of the adult population worldwide. The disease burden for hypertension is great, and it greatly increases the risk of cardiovascular morbidity and mortality. Unfortunately, there are a myriad of factors that result in an elevated blood pressure. These include genetic factors, a sedentary lifestyle, obesity, salt intake, aging, and stress. Although lifestyle modifications have had limited success, anti-hypertensive drugs have been moderately effective in lowering blood pressure. New approaches to control and treat hypertension include digital health tools and compounds that activate the angiotensin receptor type 2 (AT2), which can promote cardiovascular health. Nonetheless, research on hypertension and its management is vital for lessening the significant health and economic burden of this condition.
Collapse
Affiliation(s)
- Samaneh Goorani
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Somaye Zangene
- Faculty of Medicine, University of Tehran, Tehran 1416634793, Iran;
| | - John D. Imig
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
7
|
Amiri Roudbar M, Rosengren MK, Mousavi SF, Fegraeus K, Naboulsi R, Meadows JRS, Lindgren G. Effect of an endothelial regulatory module on plasma proteomics in exercising horses. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101265. [PMID: 38906044 DOI: 10.1016/j.cbd.2024.101265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 06/23/2024]
Abstract
Elite performing exercise requires an intricate modulation of the blood pressure to support the working muscles with oxygen. We have previously identified a genomic regulatory module that associates with differences in blood pressures of importance for elite performance in racehorses. This study aimed to determine the effect of the regulatory module on the protein repertoire. We sampled plasma from 12 Coldblooded trotters divided into two endothelial regulatory module haplotype groups, a sub-elite performing haplotype (SPH) and an elite performing haplotype (EPH), each at rest and exercise. The haplotype groups and their interaction were interrogated in two analyses, i) individual paired ratio analysis for identifying differentially abundant proteins of exercise (DAPE) and interaction (DAPI) between haplotype and exercise, and ii) unpaired ratio analysis for identifying differentially abundant protein of haplotype (DAPH). The proteomics analyses revealed a widespread change in plasma protein content during exercise, with a decreased tendency in protein abundance that is mainly related to lung function, tissue fluids, metabolism, calcium ion pathway and cellular energy metabolism. Furthermore, we provide the first investigation of the proteome variation due to the interaction between exercise and related blood pressure haplotypes, which this difference was related to a faster switch to the lipoprotein and lipid metabolism during exercise for EPH. The molecular signatures identified in the present study contribute to an improved understanding of exercise-related blood pressure regulation.
Collapse
Affiliation(s)
- Mahmoud Amiri Roudbar
- Department of Animal Science, Safiabad-Dezful Agricultural and Natural Resources Research and Education Center, Agricultural Research, Education and Extension Organization (AREEO), Dezful 333, Iran.
| | - Maria K Rosengren
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| | - Seyedeh Fatemeh Mousavi
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| | - Kim Fegraeus
- Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Sweden.
| | - Rakan Naboulsi
- Department of Women's and Children's Health, Karolinska Institute, Tomtebodavägen 18A, Stockholm 17177, Sweden.
| | - Jennifer R S Meadows
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, 75132 Uppsala, Sweden.
| | - Gabriella Lindgren
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden; Center for Animal Breeding and Genetics, Department of Biosystems, KU Leuven, 3001 Leuven, Belgium.
| |
Collapse
|
8
|
Pacella J, Lembo G, Carnevale L. A Translational Perspective on the Interplay Between Hypertension, Inflammation and Cognitive Impairment. Can J Cardiol 2024; 40:2368-2377. [PMID: 39455022 DOI: 10.1016/j.cjca.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Hypertension represents the major risk factor in the onset of cardiovascular disease worldwide. Preclinically, several mouse models of hypertension have been developed to investigate the pathophysiological link between hypertension and vascular impairment. Specifically, angiotensin-II infusion, transverse aortic constriction, deoxycorticosterone acetate salt, and N(ω)-nitro-L-arginine methyl ester (L-NAME) administration as hypertensive stimuli at the preclinical level permit the unveiling of a proinflammatory response driven by the innate and adaptive immune system and leads to vascular injury in terms of structural and functional alterations. Vascular impairment seems to be particularly critical at the cerebral level wherein arterioles, venules, and capillaries finely tune blood supply across the whole brain leading to the onset of a well known clinical condition named cerebral small vessel disease (cSVD) characterized by extensive brain injury, which culminates in the decline of cognitive functions. Advances in magnetic resonance imaging permit identification and accurate diagnosis of specific cSVD biomarkers including white matter hyperintensities, lacunar strokes, cerebral microbleeds, and enlarged perivascular spaces, each of which proved to be associated with a specific cognitive domain impairment. Such an approach in combination with pharmacological interventions targeted to the lowering of blood pressure and the prevention of vascular thrombosis formation represents a solid strategy in the prevention and the management of cSVD cognitive decay.
Collapse
Affiliation(s)
- Jacopo Pacella
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy; Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| | - Lorenzo Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy. https://twitter.com/LorCarnevale
| |
Collapse
|
9
|
Wojtacha P, Bogdańska-Chomczyk E, Majewski MK, Obremski K, Majewski MS, Kozłowska A. Renal Inflammation, Oxidative Stress, and Metabolic Abnormalities During the Initial Stages of Hypertension in Spontaneously Hypertensive Rats. Cells 2024; 13:1771. [PMID: 39513878 PMCID: PMC11545559 DOI: 10.3390/cells13211771] [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: 08/20/2024] [Revised: 10/03/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Hypertension is a major cause of mortality worldwide. The kidneys play a crucial role in regulating blood pressure and fluid volume. The relationship between the kidneys and hypertension is complex, involving factors such as the renin-angiotensin system, oxidative stress, and inflammation. This study aims to assess the levels of inflammatory markers, oxidative stress, and metabolic factors in the kidneys, focusing on their potential role in early renal damage and their association with the development of hypertension. Methods: This study was designed to compare the levels of selected inflammatory markers, e.g., interleukins, tumor necrosis factor-α (TNF-α), transforming growth factor, and serine/threonine-protein (mTOR); oxidative stress markers such as malondialdehyde, sulfhydryl group, and glucose (GLC); and metabolic markers among other enzymes, such as alanine transaminase (ALT), aspartate transaminase (AST), hexokinase II (HK-II), and hypoxia-inducible factor-1α (HIF-1α), as well as creatinine in the kidneys of spontaneously hypertensive rats (SHR/NCrl, n = 12) and Wistar Kyoto rats (WKY/NCrl, n = 12). Both juvenile (5 weeks old) and maturing (10 weeks old) specimens were examined using spectrophotometric methods, e.g., ELISA. Results: Juvenile SHRs exhibited reduced renal levels of all studied cytokines and chemokines, with lower oxidative stress and deficits in the mTOR and HK-II levels compared to the age-matched WKYs. Maturing SHRs showed increased renal levels of interleukin-1β (IL-1β), IL-6, IL-18, and TNF-α, alongside elevated carbonyl stress and increased HIF-1α as opposed to their control peers. The levels of all other studied markers were normalized in these animals, except for ALT (increased), ALP, and GLC (both reduced). Conclusions: This study underscores the significant impact of inflammatory, oxidative stress, and metabolic marker changes on renal function. Juvenile SHRs display lower marker levels, indicating an immature immune response and potential subclinical kidney damage that may contribute to hypertension development. In contrast, mature SHRs exhibit chronic inflammation, oxidative dysregulation, and metabolic disturbances, suggesting cellular damage. These changes create a feedback loop that worsens kidney function and accelerates hypertension progression, highlighting the kidneys' crucial role in both initiating and exacerbating this condition.
Collapse
Affiliation(s)
- Paweł Wojtacha
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury, Warszawska Av, 10-082 Olsztyn, Poland
| | - Ewelina Bogdańska-Chomczyk
- Department of Human Physiology and Pathophysiology, Collegium Medicum, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland; (E.B.-C.); (M.K.M.)
| | - Mariusz Krzysztof Majewski
- Department of Human Physiology and Pathophysiology, Collegium Medicum, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland; (E.B.-C.); (M.K.M.)
| | - Kazimierz Obremski
- Department of Veterinary Prevention and Feed Hygiene, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13/29, 10-718 Olsztyn, Poland;
| | - Michał Stanisław Majewski
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland;
| | - Anna Kozłowska
- Department of Human Physiology and Pathophysiology, Collegium Medicum, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland; (E.B.-C.); (M.K.M.)
| |
Collapse
|
10
|
Henriques P, Caldeira-Araújo H, Brazão MDL, Abreu AM, Vigário AM, Rosa A. Primary Dengue and Long-Term Health Status in Madeira Island, Portugal: A Retrospective Questionnaire-Based Study. Am J Trop Med Hyg 2024; 111:403-411. [PMID: 38955194 PMCID: PMC11310610 DOI: 10.4269/ajtmh.23-0502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 03/18/2024] [Indexed: 07/04/2024] Open
Abstract
Dengue is among the most important mosquito-borne viral diseases worldwide. Although its acute manifestations are well known, little is known about the long-term impact of dengue on the population's health status. Madeira Island experienced a single outbreak of autochthonous dengue from September 2012 to March 2013. To extend our knowledge about the clinical impact of the outbreak on this naive population, we applied an online questionnaire to 168 adults diagnosed with dengue at the time to characterize retrospectively their symptoms during the infection and to identify long-term manifestations, possibly triggered by dengue. The most frequent symptoms during the clinical period, reported by more than three-quarters of our participants, were fever, myalgia, extreme tiredness, and headaches, whereas vomiting, pruritus, nausea, retro-orbital pain, and arthralgia occurred in 35% to 50% of participants. In the 8 years after dengue, 61.5% of participants reported at least one recurrent previously nonexistent symptom, the most frequent being headaches, abundant hair loss, extreme tiredness, arthralgia, and myalgia, experienced by 25% to 35% of participants. Nearly 20% of the participants with persistent symptoms reported the onset of chronic illness in the 4 years after dengue, most frequently ophthalmological and autoimmune diseases (5.6% each), versus only 2.2% of chronic disease onset in participants without persistent symptoms. Our results suggest that the occurrence of persistent symptoms after primary dengue might be more frequent than anticipated and may persist for several years, having an impact on the health status and well-being of a considerable proportion of the infected population.
Collapse
Affiliation(s)
- Paulo Henriques
- Projeto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Helena Caldeira-Araújo
- Projeto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- CQM-Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Maria da Luz Brazão
- Projeto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- Serviço de Medicina Interna, Hospital Central do Funchal, SESARAM-EPERAM, Funchal, Portugal
| | - Ana Maria Abreu
- Departamento de Matemática, Faculdade de Ciências Exatas e da Engenharia, Universidade da Madeira, Funchal, Portugal
- Centro de Investigação em Matemática e Aplicações, Universidade de Évora, Évora, Portugal
| | - Ana Margarida Vigário
- Projeto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Alexandra Rosa
- Projeto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| |
Collapse
|
11
|
McDermott JG, Goodlett BL, Creed HA, Navaneethabalakrishnan S, Rutkowski JM, Mitchell BM. Inflammatory Alterations to Renal Lymphatic Endothelial Cell Gene Expression in Mouse Models of Hypertension. Kidney Blood Press Res 2024; 49:588-604. [PMID: 38972305 PMCID: PMC11345939 DOI: 10.1159/000539721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 06/02/2024] [Indexed: 07/09/2024] Open
Abstract
INTRODUCTION Hypertension (HTN) is a major cardiovascular disease that can cause and be worsened by renal damage and inflammation. We previously reported that renal lymphatic endothelial cells (LECs) increase in response to HTN and that augmenting lymphangiogenesis in the kidneys reduces blood pressure and renal pro-inflammatory immune cells in mice with various forms of HTN. Our aim was to evaluate the specific changes that renal LECs undergo in HTN. METHODS We performed single-cell RNA sequencing. Using the angiotensin II-induced and salt-sensitive mouse models of HTN, we isolated renal CD31+ and podoplanin+ cells. RESULTS Sequencing of these cells revealed three distinct cell types with unique expression profiles, including LECs. The number and transcriptional diversity of LECs increased in samples from mice with HTN, as demonstrated by 597 differentially expressed genes (p < 0.01), 274 significantly enriched pathways (p < 0.01), and 331 regulons with specific enrichment in HTN LECs. These changes demonstrate a profound inflammatory response in renal LECs in HTN, leading to an increase in genes and pathways associated with inflammation-driven growth and immune checkpoint activity in LECs. CONCLUSION These results reinforce and help to further explain the benefits of renal LECs and lymphangiogenesis in HTN.
Collapse
Affiliation(s)
- Justin G. McDermott
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | - Bethany L. Goodlett
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | - Heidi A. Creed
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | | | - Joseph M. Rutkowski
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | - Brett M. Mitchell
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| |
Collapse
|
12
|
Li Z, Xing J, Ma X, Zhang W, Wang C, Wang Y, Qi X, Liu Y, Jian D, Cheng X, Zhu Y, Shi C, Guo Y, Zhao H, Jiang W, Tang H. An orally administered bacterial membrane protein nanodrug ameliorates doxorubicin cardiotoxicity through alleviating impaired intestinal barrier. Bioact Mater 2024; 37:517-532. [PMID: 38698916 PMCID: PMC11063951 DOI: 10.1016/j.bioactmat.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/07/2024] [Accepted: 03/20/2024] [Indexed: 05/05/2024] Open
Abstract
The cardiotoxicity caused by Dox chemotherapy represents a significant limitation to its clinical application and is a major cause of late death in patients undergoing chemotherapy. Currently, there are no effective treatments available. Our analysis of 295 clinical samples from 132 chemotherapy patients and 163 individuals undergoing physical examination revealed a strong positive correlation between intestinal barrier injury and the development of cardiotoxicity in chemotherapy patients. We developed a novel orally available and intestinal targeting protein nanodrug by assembling membrane protein Amuc_1100 (obtained from intestinal bacteria Akkermansia muciniphila), fluorinated polyetherimide, and hyaluronic acid. The protein nanodrug demonstrated favorable stability against hydrolysis compared with free Amuc_1100. The in vivo results demonstrated that the protein nanodrug can alleviate Dox-induced cardiac toxicity by improving gut microbiota, increasing the proportion of short-chain fatty acid-producing bacteria from the Lachnospiraceae family, and further enhancing the levels of butyrate and pentanoic acids, ultimately regulating the homeostasis repair of lymphocytes in the spleen and heart. Therefore, we believe that the integrity of the intestinal barrier plays an important role in the development of chemotherapy-induced cardiotoxicity. Protective interventions targeting the intestinal barrier may hold promise as a general clinical treatment regimen for reducing Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhen Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Junyue Xing
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xiaohan Ma
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Wanjun Zhang
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Chuan Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Yingying Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xinkun Qi
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Yanhui Liu
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Dongdong Jian
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xiaolei Cheng
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Yanjie Zhu
- Department of Pathology, Central Hospital of Kaifeng City, Kaifeng, Henan, 475000, China
| | - Chao Shi
- Henan Key Laboratory of Molecular Pathology, Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Yongjun Guo
- Henan Key Laboratory of Molecular Pathology, Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Huan Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Jiang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Hao Tang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| |
Collapse
|
13
|
Peter JK, Umene R, Wu CH, Nakamura Y, Washimine N, Yamamoto R, Ngugi C, Linge K, Kweri JK, Inoue T. Renal macrophages induce hypertension and kidney fibrosis in Angiotensin II salt mice model. Biochem Biophys Res Commun 2024; 715:149997. [PMID: 38678782 DOI: 10.1016/j.bbrc.2024.149997] [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: 01/15/2024] [Revised: 04/12/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
The immune system is involved in hypertension development with different immune cells reported to have either pro or anti-hypertensive effects. In hypertension, immune cells have been thought to infiltrate blood pressure-regulating organs, resulting in either elevation or reduction of blood pressure. There is controversy over whether macrophages play a detrimental or beneficial role in the development of hypertension, and the few existing studies have yielded conflicting results. This study aimed to determine the effects of angiotensin II (Ang II) salt-induced hypertension on renal immune cells and to determine whether renal macrophages are involved in the induction of hypertension. Hypertension was induced by administration of Ang II and saline for two weeks. The effects of hypertension on kidney immune cells were assessed using flow cytometry. Macrophage infiltration in the kidney was assessed by immunohistochemistry and kidney fibrosis was assessed using trichrome stain and kidney real time-qPCR. Liposome encapsulated clodronate was used to deplete macrophages in C57BL/6J mice and investigate the direct role of macrophages in hypertension induction. Ang II saline mice group developed hypertension, had increased renal macrophages, and had increased expression of Acta2 and Col1a1 and kidney fibrotic areas. Macrophage depletion blunted hypertension development and reduced the expression of Acta2 and Col1a1 in the kidney and kidney fibrotic areas in Ang II saline group. The results of this study demonstrate that macrophages infiltrate the kidneys and increase kidney fibrosis in Ang II salt-induced hypertension, and depletion of macrophages suppresses the development of hypertension and decreases kidney fibrosis. This indicates that macrophages play a direct role in hypertension development. Hence macrophages have a potential to be considered as therapeutic target in hypertension management.
Collapse
Affiliation(s)
- Joseph Kasyoki Peter
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan; Department of Medical Physiology, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya; Department of Clinical Medicine, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Ryusuke Umene
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan; Department of Nephrology, Graduate School of Biomedical Sciences, Nagasaki University, Japan.
| | - Chia-Hsien Wu
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Yasuna Nakamura
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Norito Washimine
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan; Department of Nephrology, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Ryoko Yamamoto
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Caroline Ngugi
- Department of Medical Microbiology, School of Biomedical Sciences, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Kavoo Linge
- Department of Medical Physiology, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Joseph K Kweri
- Department of Human Anatomy, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Tsuyoshi Inoue
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan.
| |
Collapse
|
14
|
Prakash P, Swami Vetha BS, Chakraborty R, Wenegieme TY, Masenga SK, Muthian G, Balasubramaniam M, Wanjalla CN, Hinton AO, Kirabo A, Williams CR, Aileru A, Dash C. HIV-Associated Hypertension: Risks, Mechanisms, and Knowledge Gaps. Circ Res 2024; 134:e150-e175. [PMID: 38781298 PMCID: PMC11126208 DOI: 10.1161/circresaha.124.323979] [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] [Indexed: 05/25/2024]
Abstract
HIV type 1 (HIV-1) is the causative agent of AIDS. Since the start of the epidemic, HIV/AIDS has been responsible for ≈40 million deaths. Additionally, an estimated 39 million people are currently infected with the virus. HIV-1 primarily infects immune cells, such as CD4+ (cluster of differentiation 4+) T lymphocytes (T cells), and as a consequence, the number of CD4+ T cells progressively declines in people living with HIV. Within a span of ≈10 years, HIV-1 infection leads to the systemic failure of the immune system and progression to AIDS. Fortunately, potent antiviral therapy effectively controls HIV-1 infection and prevents AIDS-related deaths. The efficacy of the current antiviral therapy regimens has transformed the outcome of HIV/AIDS from a death sentence to a chronic disease with a prolonged lifespan of people living with HIV. However, antiviral therapy is not curative, is challenged by virus resistance, can be toxic, and, most importantly, requires lifelong adherence. Furthermore, the improved lifespan has resulted in an increased incidence of non-AIDS-related morbidities in people living with HIV including cardiovascular diseases, renal disease, liver disease, bone disease, cancer, and neurological conditions. In this review, we summarize the current state of knowledge of the cardiovascular comorbidities associated with HIV-1 infection, with a particular focus on hypertension. We also discuss the potential mechanisms known to drive HIV-1-associated hypertension and the knowledge gaps in our understanding of this comorbid condition. Finally, we suggest several directions of future research to better understand the factors, pathways, and mechanisms underlying HIV-1-associated hypertension in the post-antiviral therapy era.
Collapse
Affiliation(s)
- Prem Prakash
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Berwin Singh Swami Vetha
- Department of Foundational Sciences and Research, School of Dental Medicine, East Carolina University, 1851 MacGregor Downs Road, MS 701, Greenville, NC 27834
| | - Rajasree Chakraborty
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Tara-Yesomi Wenegieme
- Department of Neuroscience, Cell Biology and Physiology; Boonshoft School of Medicine and the College of Science and Mathematics; Wright State University, Dayton, OH 45435, USA
| | - Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Kabwe, Central Province, 10101, Zambia
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Kabwe, Central Province, 10101, Zambia
| | - Gladson Muthian
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Muthukumar Balasubramaniam
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | | | - Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology; Boonshoft School of Medicine and the College of Science and Mathematics; Wright State University, Dayton, OH 45435, USA
| | - Azeez Aileru
- Department of Foundational Sciences and Research, School of Dental Medicine, East Carolina University, 1851 MacGregor Downs Road, MS 701, Greenville, NC 27834
| | - Chandravanu Dash
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| |
Collapse
|
15
|
Hao W, Liu W, Chang R, Yang M, Xin K, Liu J, Wang Y, Ren M, Xie J, Yang Y. Safety and clinical efficacy of immune checkpoint inhibitors in advanced gastric cancer in the real world. J Cancer Res Clin Oncol 2024; 150:180. [PMID: 38587528 PMCID: PMC11001672 DOI: 10.1007/s00432-024-05703-8] [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: 01/27/2024] [Accepted: 03/14/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND To evaluate the clinical efficacy and safety of immune checkpoint inhibitors in patients with advanced gastric cancer in the real world. METHODS The retrospective analysis was conducted on the clinical records of 402 patients with advanced gastric cancer who were admitted to the Nanjing Drum Tower Hospital between December 2017 and April 2022 and who had received immunotherapy. Observation target: drug use, treatment, adverse reaction type and grade, objective response rate (ORR), disease control rate (DCR), progression free survival (PFS), and overall survival (OS). RESULTS By retrospectively analyzing the data of patients with advanced gastric cancer treated with ICIs previously admitted to our medical center, we found some clinical characteristic factors associated with the occurrence of irAEs as well as the efficacy and prognosis: the presence or absence of hypertension, whether or not to receive targeted therapies can predict the occurrence of immune-related adverse events (irAEs), and the more the presence of irAEs, the better the prognosis. These can help clinicians in clinical drug selection. CONCLUSIONS The results of this paper show that the occurrence of irAEs is associated with patients' OS. irAEs occurrence can prolong patients' OS. irAEs occurrence may serve as a surrogate marker for ICIs.
Collapse
Affiliation(s)
- Wen Hao
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenjing Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ruimin Chang
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mi Yang
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Kai Xin
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingxin Liu
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yibing Wang
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meijin Ren
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiaqi Xie
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Yang Yang
- Department of Oncology, Nanjing Drum Tower Hospital, Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
16
|
Moss A, Kuttippurathu L, Srivastava A, Schwaber JS, Vadigepalli R. Dynamic dysregulation of transcriptomic networks in brainstem autonomic nuclei during hypertension development in the female spontaneously hypertensive rat. Physiol Genomics 2024; 56:283-300. [PMID: 38145287 PMCID: PMC11283910 DOI: 10.1152/physiolgenomics.00073.2023] [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: 07/21/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023] Open
Abstract
Neurogenic hypertension stems from an imbalance in autonomic function that shifts the central cardiovascular control circuits toward a state of dysfunction. Using the female spontaneously hypertensive rat and the normotensive Wistar-Kyoto rat model, we compared the transcriptomic changes in three autonomic nuclei in the brainstem, nucleus of the solitary tract (NTS), caudal ventrolateral medulla, and rostral ventrolateral medulla (RVLM) in a time series at 8, 10, 12, 16, and 24 wk of age, spanning the prehypertensive stage through extended chronic hypertension. RNA-sequencing data were analyzed using an unbiased, dynamic pattern-based approach that uncovered dominant and several subtle differential gene regulatory signatures. Our results showed a persistent dysregulation across all three autonomic nuclei regardless of the stage of hypertension development as well as a cascade of transient dysregulation beginning in the RVLM at the prehypertensive stage that shifts toward the NTS at the hypertension onset. Genes that were persistently dysregulated were heavily enriched for immunological processes such as antigen processing and presentation, the adaptive immune response, and the complement system. Genes with transient dysregulation were also largely region-specific and were annotated for processes that influence neuronal excitability such as synaptic vesicle release, neurotransmitter transport, and an array of neuropeptides and ion channels. Our results demonstrate that neurogenic hypertension is characterized by brainstem region-specific transcriptomic changes that are highly dynamic with significant gene regulatory changes occurring at the hypertension onset as a key time window for dysregulation of homeostatic processes across the autonomic control circuits.NEW & NOTEWORTHY Hypertension is a major disease and is the primary risk factor for cardiovascular complications and stroke. The gene expression changes in the central nervous system circuits driving hypertension are understudied. Here, we show that coordinated and region-specific gene expression changes occur in the brainstem autonomic circuits over time during the development of a high blood pressure phenotype in a rat model of human essential hypertension.
Collapse
Affiliation(s)
- Alison Moss
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Lakshmi Kuttippurathu
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Ankita Srivastava
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - James S Schwaber
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
17
|
Lu Y, Luo Q, Liu Y, Wang H. Relationships between inflammation markers and the risk of hypertension in primary Sjögren's syndrome: A retrospective cohort study. Mod Rheumatol 2024; 34:369-375. [PMID: 36976576 DOI: 10.1093/mr/road032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/01/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
OBJECTIVES The association of inflammation markers with hypertension (HTN) in primary Sjögren's syndrome (pSS) remains controversial. We aimed to investigate whether inflammation markers are at increased risk of developing HTN in pSS patients. METHODS A retrospective cohort study included pSS patients (n = 380) between May 2011 and May 2020 from the Third People's Hospital of Chengdu. Multivariable Cox regression analyses were used to estimate hazard ratios (HRs) of the potential inflammation markers for pSS-HTN. Subsequently, the dose-response relationships were also used. RESULTS Out of 380 pSS patients, 171 (45%) developed HTN, and the median follow-up period was 4.16 years. Univariable Cox regression analysis showed that the erythrocyte sedimentation rate (ESR) and neutrophils were significantly associated with the incident HTN (P < 0.05). After adjustment for covariates, this association between ESR (adjusted HR 1.017, 95%CI: 1.005-1.027, P = .003), neutrophils (adjusted HR 1.356, 95%CI: 1.113-1.653, P = .003), and HTN remained significant. The dose-effect relationship was also found between ESR, neutrophils, and HTN (P = .001). CONCLUSIONS Inflammation markers may play an important role in the incident HTN in pSS.
Collapse
Affiliation(s)
- Yan Lu
- Department of Cardiology, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
- Department of Geriatrics, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qiang Luo
- Department of Cardiology, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| | - Yaping Liu
- Department of Cardiology, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
- Department of Geriatrics, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Han Wang
- Department of Cardiology, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| |
Collapse
|
18
|
Skiba D, Jaskuła K, Nawrocka A, Poznański P, Łazarczyk M, Szymański Ł, Żera T, Sacharczuk M, Cudnoch-Jędrzejewska A, Gaciong Z. The Role of Opioid Receptor Antagonists in Regulation of Blood Pressure and T-Cell Activation in Mice Selected for High Analgesia Induced by Swim Stress. Int J Mol Sci 2024; 25:2618. [PMID: 38473865 DOI: 10.3390/ijms25052618] [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: 01/17/2024] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
Opioid peptides and their G protein-coupled receptors are important regulators within the cardiovascular system, implicated in the modulation of both heart and vascular functions. It is known that naloxone-an opioid antagonist-may exert a hypertensive effect. Recent experimental and clinical evidence supports the important role of inflammatory mechanisms in hypertension. Since opioids may play a role in the regulation of both blood pressure and immune response, we studied these two processes in our model. We aimed to evaluate the effect of selective and non-selective opioid receptor antagonists on blood pressure and T-cell activation in a mouse model of high swim stress-induced analgesia. Blood pressure was measured before and during the infusion of opioid receptor antagonists using a non-invasive tail-cuff measurement system. To assess the activation of T-cells, flow cytometry was used. We discovered that the non-selective antagonism of the opioid system by naloxone caused a significant elevation of blood pressure. The selective antagonism of μ and κ but not δ opioid receptors significantly increased systolic blood pressure. Subsequently, a brief characterization of T-cell subsets was performed. We found that the blockade of μ and δ receptors is associated with the increased expression of CD69 on CD4 T-cells. Moreover, we observed an increase in the central memory CD4 and central memory CD8 T-cell populations after the δ opioid receptor blockade. The antagonism of the μ opioid receptor increased the CD8 effector and central memory T-cell populations.
Collapse
Affiliation(s)
- Dominik Skiba
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A Street, Jastrzebiec, 05-552 Magdalenka, Poland
| | - Kinga Jaskuła
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A Street, Jastrzebiec, 05-552 Magdalenka, Poland
| | - Agata Nawrocka
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A Street, Jastrzebiec, 05-552 Magdalenka, Poland
| | - Piotr Poznański
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A Street, Jastrzebiec, 05-552 Magdalenka, Poland
| | - Marzena Łazarczyk
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A Street, Jastrzebiec, 05-552 Magdalenka, Poland
| | - Łukasz Szymański
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A Street, Jastrzebiec, 05-552 Magdalenka, Poland
| | - Tymoteusz Żera
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Banacha 1B Street, 02-097 Warsaw, Poland
| | - Mariusz Sacharczuk
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A Street, Jastrzebiec, 05-552 Magdalenka, Poland
- Department of Pharmacodynamics, Medical University of Warsaw, Zwirki i Wigury 81 Street, 02-091 Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Banacha 1B Street, 02-097 Warsaw, Poland
| | - Zbigniew Gaciong
- Department and Clinic of Internal Diseases, Hypertension and Angiology, Medical University of Warsaw, Banacha 1A Street, 02-097 Warsaw, Poland
| |
Collapse
|
19
|
Perrotta S, Carnevale D. Brain-Splenic Immune System Interactions in Hypertension: Cellular and Molecular Mechanisms. Arterioscler Thromb Vasc Biol 2024; 44:65-75. [PMID: 37942610 DOI: 10.1161/atvbaha.123.318230] [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: 04/07/2023] [Accepted: 10/20/2023] [Indexed: 11/10/2023]
Abstract
Hypertension represents a major worldwide cause of death and disability, and it is becoming increasingly clear that available therapies are not sufficient to reduce the risk of major cardiovascular events. Various mechanisms contribute to blood pressure increase: neurohormonal activation, autonomic nervous system imbalance, and immune activation. Of note, the brain is an important regulator of blood pressure levels; it recognizes the peripheral perturbation and organizes a reflex response by modulating immune system and hormonal release to attempt at restoring the homeostasis. The connection between the brain and peripheral organs is mediated by the autonomic nervous system, which also modulates immune and inflammatory responses. Interestingly, an increased autonomic nervous system activity has been correlated with an altered immune response in cardiovascular diseases. The spleen is the largest immune organ exerting a potent influence on the cardiovascular system during disease and is characterized by a dense noradrenergic innervation. Taken together, these aspects led to hypothesize a key role of neuroimmune mechanisms in the onset and progression of hypertension. This review discusses how the nervous and splenic immune systems interact and how the mechanisms underlying the neuroimmune cross talk influence the disease progression.
Collapse
Affiliation(s)
- Sara Perrotta
- Department of Angiocardioneurology and Translational Medicine, Unit of Neuro and Cardiovascular Pathophysiology, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, Pozzilli, Italy (S.P., D.C.)
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, Unit of Neuro and Cardiovascular Pathophysiology, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, Pozzilli, Italy (S.P., D.C.)
- Department of Molecular Medicine, "Sapienza" University of Rome, Italy (D.C.)
| |
Collapse
|
20
|
Vallianou NG, Kounatidis D, Panagopoulos F, Evangelopoulos A, Stamatopoulos V, Papagiorgos A, Geladari E, Dalamaga M. Gut Microbiota and Its Role in the Brain-Gut-Kidney Axis in Hypertension. Curr Hypertens Rep 2023; 25:367-376. [PMID: 37632662 DOI: 10.1007/s11906-023-01263-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2023] [Indexed: 08/28/2023]
Abstract
PURPOSE OF REVIEW The role of the gut microbiota in modulating blood pressure is increasingly being recognized, currently. The purpose of this review is to summarize recent findings about the mechanisms involved in hypertension with regard to the phenomenon of "gut dysbiosis." RECENT FINDINGS Gut dysbiosis, i.e., the imbalance between the gut microbiota and the host, is characterized by a disruption of the tight junction proteins, such as occludins, claudins, and JAMs (junctional adhesion molecules), resulting in increased gut permeability or the so called "leaky gut." Due to the influence of genetic as well as environmental factors, various metabolites produced by the gut microbiota, such as indole and p-cresol, are increased. Thereby, uremic toxins, such as indoxyl sulfates and p-cresol sulfates, accumulate in the blood and the urine, causing damage in the podocytes and the tubular cells. In addition, immunological mechanisms are implicated as well. In particular, a switch from M2 macrophages to M1 macrophages, which produce pro-inflammatory cytokines, occurs. Moreover, a higher level of Th17 cells, releasing large amounts of interleukin-17 (IL-17), has been reported, when a diet rich in salt is consumed. Therefore, apart from the aggravation of uremic toxins, which may account for direct harmful effects on the kidney, there is inflammation not only in the gut, but in the kidneys as well. This crosstalk between the gut and the kidney is suggested to play a crucial role in hypertension. Notably, the brain is also implicated, with an increasing sympathetic output. The brain-gut-kidney axis seems to be deeply involved in the development of hypertension and chronic kidney disease (CKD). The notion that, by modulating the gut microbiota, we could regulate blood pressure is strongly supported by the current evidence. A healthy diet, low in animal protein and fat, and low in salt, together with the utilization of probiotics, prebiotics, synbiotics, or postbiotics, may contribute to our fight against hypertension.
Collapse
Affiliation(s)
| | | | - Fotis Panagopoulos
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | | | | | | | - Eleni Geladari
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, Athens, Greece
| |
Collapse
|
21
|
Kaplan A, Lakkis B, El-Samadi L, Karaayvaz EB, Booz GW, Zouein FA. Cooling Down Inflammation in the Cardiovascular System via the Nicotinic Acetylcholine Receptor. J Cardiovasc Pharmacol 2023; 82:241-265. [PMID: 37539950 DOI: 10.1097/fjc.0000000000001455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023]
Abstract
ABSTRACT Inflammation is a major player in many cardiovascular diseases including hypertension, atherosclerosis, myocardial infarction, and heart failure. In many individuals, these conditions coexist and mutually exacerbate each other's progression. The pathophysiology of these diseases entails the active involvement of both innate and adaptive immune cells. Immune cells that possess the α7 subunit of the nicotinic acetylcholine receptor on their surface have the potential to be targeted through both pharmacological and electrical stimulation of the cholinergic system. The cholinergic system regulates the inflammatory response to various stressors in different organ systems by systematically suppressing spleen-derived monocytes and chemokines and locally improving immune cell function. Research on the cardiovascular system has demonstrated the potential for atheroma plaque stabilization and regression as favorable outcomes. Smaller infarct size and reduced fibrosis have been associated with improved cardiac function and a decrease in adverse cardiac remodeling. Furthermore, enhanced electrical stability of the myocardium can lead to a reduction in the incidence of ventricular tachyarrhythmia. In addition, improving mitochondrial dysfunction and decreasing oxidative stress can result in less myocardial tissue damage caused by reperfusion injury. Restoring baroreflex activity and reduction in renal damage can promote blood pressure regulation and help counteract hypertension. Thus, the present review highlights the potential of nicotinic acetylcholine receptor activation as a natural approach to alleviate the adverse consequences of inflammation in the cardiovascular system.
Collapse
Affiliation(s)
- Abdullah Kaplan
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
- Department of Cardiology, Kemer Public Hospital, Kemer, Antalya, Turkey
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
| | - Bachir Lakkis
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
| | - Lana El-Samadi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
| | - Ekrem Bilal Karaayvaz
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS; and
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS; and
- Department of Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Inserm, Université Paris-Saclay, France
| |
Collapse
|
22
|
Kanclerska J, Szymańska-Chabowska A, Poręba R, Michałek-Zrąbkowska M, Lachowicz G, Mazur G, Martynowicz H. A Systematic Review of Publications on the Associations Between Sleep Architecture and Arterial Hypertension. Med Sci Monit 2023; 29:e941066. [PMID: 37665688 PMCID: PMC10487188 DOI: 10.12659/msm.941066] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/22/2023] [Indexed: 09/06/2023] Open
Abstract
Sleep research has garnered substantial interest among scientists owing to its correlation with various diseases, particularly elevated blood pressure observed in patients with obstructive sleep apnea. This systematic review aims to identify and analyze publications exploring the associations between sleep architecture and arterial hypertension. A comprehensive search of PubMed (MEDLINE), Scopus, and Embase databases yielded 111 reports, of which 7 manuscripts were included in the review. Four of the studies reported a significant reduction in the duration of the N3 phase of sleep in hypertensive patients, while 2 studies found a statistically significant reduction in the duration of the N2 and rapid eye movement (REM) stages of sleep. Three studies indicated increased sleep fragmentation in hypertensive patients. They showed a longer duration of the N1 stage of sleep, shorter duration of overall sleep time, and an increased apnea-hypopnea index in hypertensive patients. These findings underscore the association between the duration of non-REM/REM sleep stages and elevated BP, providing substantial evidence. Moreover, a notable increase in sleep fragmentation was observed among patients with hypertension. However, further research is warranted to expand and deepen our understanding of this intricate relationship. This systematic review serves as a valuable resource, guiding future investigations and contributing to advancements in the field of sleep and arterial hypertension.
Collapse
|
23
|
Dai Y, Shen Z, Khachatryan LG, Vadiyan DE, Karampoor S, Mirzaei R. Unraveling mechanistic insights into the role of microbiome in neurogenic hypertension: A comprehensive review. Pathol Res Pract 2023; 249:154740. [PMID: 37567034 DOI: 10.1016/j.prp.2023.154740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023]
Abstract
Neurogenic hypertension, a complex and multifactorial cardiovascular disorder, is known to be influenced by various genetic, environmental, and lifestyle factors. In recent years, there has been growing interest in the role of the gut microbiome in hypertension pathogenesis. The bidirectional communication between the gut microbiota and the central nervous system, known as the microbiota-gut-brain axis, has emerged as a crucial mechanism through which the gut microbiota exerts its influence on neuroinflammation, immune responses, and blood pressure regulation. Recent studies have shown how the microbiome has a substantial impact on a variety of physiological functions, such as cardiovascular health. The increased sympathetic activity to the gut may cause microbial dysbiosis, increased permeability of the gut, and increased inflammatory reactions by altering a number of intestinal bacteria producing short-chain fatty acids (SCFAs) and the concentrations of lipopolysaccharide (LPS) in the plasma. Collectively, these microbial metabolic and structural compounds stimulate sympathetic stimulation, which may be an important stage in the onset of hypertension. The result is an upsurge in peripheral and central inflammatory response. In addition, it has recently been shown that a link between the immune system and the gut microbiota might play a significant role in hypertension. The therapeutic implications of the gut microbiome including probiotic usage, prebiotics, dietary modifications, and fecal microbiota transplantation in neurogenic hypertension have also been found. A large body of research suggests that probiotic supplementation might help reduce chronic inflammation and hypertension that have an association with dysbiosis in the gut microbiota. Overall, this review sheds light on the intricate interplay between the gut microbiome and neurogenic hypertension, providing valuable insights for both researchers and clinicians. As our knowledge of the microbiome's role in hypertension expands, novel therapeutic strategies and diagnostic biomarkers may pave the way for more effective management and prevention of this prevalent cardiovascular disorder. Exploring the potential of the microbiome in hypertension offers an exciting avenue for future research and offers opportunities for precision medicine and improved patient care.
Collapse
Affiliation(s)
- Yusang Dai
- Physical Examination Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Zheng Shen
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Lusine G Khachatryan
- Department of Pediatric Diseases, N.F. Filatov Clinical Institute of Children's Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Russia
| | - Diana E Vadiyan
- Institute of Dentistry, Department of Pediatric, Preventive Dentistry and Orthodontics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Russia
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
24
|
Soegiarto G, Mahdi BA, Wulandari L, Fahmita KD, Hadmoko ST, Gautama HI, Prasetyaningtyas D, Prasetyo ME, Negoro PP, Arafah N, Purnomosari D, Tinduh D, Husada D, Baskoro A, Fetarayani D, Nurani WK, Oceandy D. Evaluation of Antibody Response and Adverse Effects following Heterologous COVID-19 Vaccine Booster with mRNA Vaccine among Healthcare Workers in Indonesia. Vaccines (Basel) 2023; 11:1160. [PMID: 37514976 PMCID: PMC10386191 DOI: 10.3390/vaccines11071160] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Background: The administration of the third (or booster) dose of COVID-19 vaccine is important in maintaining protection against SARS-CoV-2 infection or the severity of the disease. In Indonesia, health care workers (HCWs) are among the first to receive a booster dose of the COVID-19 vaccine. In this study, we evaluated the antibody response and adverse events following heterologous booster vaccine using mRNA-1273 among HCWs that were fully vaccinated with inactivated viral vaccine as the priming doses. Methods: 75 HCWs at Dr. Soetomo General Hospital in Surabaya, Indonesia, participated in this study. The level of antibody against the SARS-CoV-2 receptor binding domain was analyzed at 1, 3, and 5 months following the second priming dose and at 1, 3, and 5 months after the booster dose. Results: We found a significantly higher level of antibody response in subjects receiving a booster dose of the mRNA-1273 vaccine compared to those receiving an inactivated viral vaccine as a booster. Interestingly, participants with hypertension and a history of diabetes mellitus showed a lower antibody response following the booster dose. There was a higher frequency of adverse events following injection with the mRNA-1273 vaccine compared to the inactivated viral vaccine, although the overall adverse events were considered minor. Conclusions: A heterologous booster dose using mRNA vaccine resulted in a high antibody response; however, participants with hypertension and diabetes mellitus displayed a lower antibody response.
Collapse
Affiliation(s)
- Gatot Soegiarto
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
- Postgraduate School, Master Program on Immunology, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Bagus Aulia Mahdi
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Laksmi Wulandari
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Karin Dhia Fahmita
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Satrio Tri Hadmoko
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Hendra Ikhwan Gautama
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Dewi Prasetyaningtyas
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Muhammad Edwin Prasetyo
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Pujo Prawiro Negoro
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Nur Arafah
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Dewajani Purnomosari
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Damayanti Tinduh
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Dominicus Husada
- Department of Child Health, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Ari Baskoro
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Deasy Fetarayani
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Wita Kartika Nurani
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60132, Indonesia
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
- Department of Biomedical Science, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| |
Collapse
|
25
|
Rivero-Pino F, Villanueva Á, Montserrat-de-la-Paz S, Sanchez-Fidalgo S, Millán-Linares MC. Evidence of Immunomodulatory Food-Protein Derived Peptides in Human Nutritional Interventions: Review on the Outcomes and Potential Limitations. Nutrients 2023; 15:2681. [PMID: 37375585 DOI: 10.3390/nu15122681] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The immune system is somehow related to all the metabolic pathways, in a bidirectional way, and the nutritional interventions affecting these pathways might have a relevant impact on the inflammatory status of the individuals. Food-derived peptides have been demonstrated to exert several bioactivities by in vitro or animal studies. Their potential to be used as functional food is promising, considering the simplicity of their production and the high value of the products obtained. However, the number of human studies performed until now to demonstrate effects in vivo is still scarce. Several factors must be taken into consideration to carry out a high-quality human study to demonstrate immunomodulatory-promoting properties of a test item. This review aims to summarize the recent human studies published in which the purpose was to demonstrate bioactivity of protein hydrolysates, highlighting the main results and the limitations that can restrict the relevance of the studies. Results collected are promising, although in some studies, physiological changes could not be observed. When responses were observed, they sometimes did not refer to relevant parameters and the immunomodulatory properties could not be clearly established with the current evidence. Well-designed clinical trials are needed in order to evaluate the role of protein hydrolysates in immunonutrition.
Collapse
Affiliation(s)
- Fernando Rivero-Pino
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Álvaro Villanueva
- Department of Food & Health, Instituto de la Grasa (IG-CSIC), Campus Universitario Pablo de Olavide, Ctra. Utrera Km. 1, 41013 Seville, Spain
| | - Sergio Montserrat-de-la-Paz
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Susana Sanchez-Fidalgo
- Department of Preventive Medicine and Public Health, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Maria C Millán-Linares
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| |
Collapse
|
26
|
Chen Y, Liu Z, Gong Y. Neuron-immunity communication: mechanism of neuroprotective effects in EGCG. Crit Rev Food Sci Nutr 2023; 64:9333-9352. [PMID: 37216484 DOI: 10.1080/10408398.2023.2212069] [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] [Indexed: 05/24/2023]
Abstract
Epigallocatechin gallate (EGCG), a naturally occurring active ingredient unique to tea, has been shown to have neuroprotective potential. There is growing evidence of its potential advantages in the prevention and treatment of neuroinflammation, neurodegenerative diseases, and neurological damage. Neuroimmune communication is an important physiological mechanism in neurological diseases, including immune cell activation and response, cytokine delivery. EGCG shows great neuroprotective potential by modulating signals related to autoimmune response and improving communication between the nervous system and the immune system, effectively reducing the inflammatory state and neurological function. During neuroimmune communication, EGCG promotes the secretion of neurotrophic factors into the repair of damaged neurons, improves intestinal microenvironmental homeostasis, and ameliorates pathological phenotypes through molecular and cellular mechanisms related to the brain-gut axis. Here, we discuss the molecular and cellular mechanisms of inflammatory signaling exchange involving neuroimmune communication. We further emphasize that the neuroprotective role of EGCG is dependent on the modulatory role between immunity and neurology in neurologically related diseases.
Collapse
Affiliation(s)
- Ying Chen
- Key Laboratory of Tea Science of Ministry of Educatioxn, Changsha, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, China
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Educatioxn, Changsha, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha, China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, China
| | - Yushun Gong
- Key Laboratory of Tea Science of Ministry of Educatioxn, Changsha, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, China
| |
Collapse
|
27
|
Wang X, Wu X, Zhang P, Zhou Y, Cai J, Jin L. Single-cell transcriptome profiling reveals enriched memory T-cell subpopulations in hypertension. Front Cell Dev Biol 2023; 11:1132040. [PMID: 37009484 PMCID: PMC10060952 DOI: 10.3389/fcell.2023.1132040] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction: The adaptive immune response mediated by T cells plays a vital role in the initiation and maintenance of blood pressure (BP) elevation. Memory T cells, which are antigen-specific T cells, can respond specifically to repeated hypertensive stimuli. Although the roles of memory T cells in animal models are well studied, their maintenance and functions in hypertensive patients are poorly understood.Method: Here, we focused on the circulating memory T cells of hypertensive patients. By using single-cell RNA sequencing technology, subsets of memory T cells were identified. Differentially expressed genes (DEGs) and functional pathways were explored for related biological functions in each population of memory T cells.Result and Discussion: Our study identified four subsets of memory T cells in the blood of hypertensive patients, with CD8 effector memory T (TEM) cells accounting for more cells and demonstrating more biological functions than CD4 TEM cells. CD8 TEM cells were further analyzed using single-cell RNA sequencing technology, and subpopulation 1 was demonstrated to contribute to BP elevation. The key marker genes CKS2, PLIN2, and CNBP were identified and validated by mass-spectrum flow cytometry. Our data suggest that CD8 TEM cells as well as the marker genes could be preventive targets for patients with hypertensive cardiovascular disease.
Collapse
Affiliation(s)
- Xiaoqi Wang
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, China
| | - Xiaobin Wu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Ministry of Education Key Laboratory of Molecular Cardiovascular Sciences, Peking University, Beijing, China
| | - Pei Zhang
- Department of Hypertension, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Henan, China
| | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Ministry of Education Key Laboratory of Molecular Cardiovascular Sciences, Peking University, Beijing, China
- *Correspondence: Yuan Zhou, ; Jun Cai, ; Ling Jin,
| | - Jun Cai
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, China
- *Correspondence: Yuan Zhou, ; Jun Cai, ; Ling Jin,
| | - Ling Jin
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- *Correspondence: Yuan Zhou, ; Jun Cai, ; Ling Jin,
| |
Collapse
|
28
|
Omit SBS, Akhter S, Rana HK, Rana ARMMH, Podder NK, Rakib MI, Nobi A. Identification of Comorbidities, Genomic Associations, and Molecular Mechanisms for COVID-19 Using Bioinformatics Approaches. BIOMED RESEARCH INTERNATIONAL 2023; 2023:6996307. [PMID: 36685671 PMCID: PMC9848821 DOI: 10.1155/2023/6996307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/09/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023]
Abstract
Several studies have been done to identify comorbidities of COVID-19. In this work, we developed an analytical bioinformatics framework to reveal COVID-19 comorbidities, their genomic associations, and molecular mechanisms accomplishing transcriptomic analyses of the RNA-seq datasets provided by the Gene Expression Omnibus (GEO) database, where normal and infected tissues were evaluated. Using the framework, we identified 27 COVID-19 correlated diseases out of 7,092 collected diseases. Analyzing clinical and epidemiological research, we noticed that our identified 27 diseases are associated with COVID-19, where hypertension, diabetes, obesity, and lung cancer are observed several times in COVID-19 patients. Therefore, we selected the above four diseases and performed assorted analyses to demonstrate the association between COVID-19 and hypertension, diabetes, obesity, and lung cancer as comorbidities. We investigated genomic associations with the cross-comparative analysis and Jaccard's similarity index, identifying shared differentially expressed genes (DEGs) and linking DEGs of COVID-19 and the comorbidities, in which we identified hypertension as the most associated illness. We also revealed molecular mechanisms by identifying statistically significant ten pathways and ten ontologies. Moreover, to understand cellular physiology, we did protein-protein interaction (PPI) analyses among the comorbidities and COVID-19. We also used the degree centrality method and identified ten biomarker hub proteins (IL1B, CXCL8, FN1, MMP9, CXCL10, IL1A, IRF7, VWF, CXCL9, and ISG15) that associate COVID-19 with the comorbidities. Finally, we validated our findings by searching the published literature. Thus, our analytical approach elicited interconnections between COVID-19 and the aforementioned comorbidities in terms of remarkable DEGs, pathways, ontologies, PPI, and biomarker hub proteins.
Collapse
Affiliation(s)
- Shudeb Babu Sen Omit
- Department of Computer Science and Telecommunication Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Salma Akhter
- Department of Environmental Science and Disaster Management, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Humayan Kabir Rana
- Department of Computer Science and Engineering, Green University of Bangladesh, Dhaka 1207, Bangladesh
| | - A. R. M. Mahamudul Hasan Rana
- Department of Computer Science and Telecommunication Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Nitun Kumar Podder
- Department of Computer Science and Engineering, Khulna University of Engineering & Technology, Khulna 9203, Bangladesh
| | - Mahmudul Islam Rakib
- Department of Computer Science and Telecommunication Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Ashadun Nobi
- Department of Computer Science and Telecommunication Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| |
Collapse
|
29
|
Kinin B1 Receptor Mediates Bidirectional Interaction between Neuroinflammation and Oxidative Stress. Antioxidants (Basel) 2023; 12:antiox12010150. [PMID: 36671012 PMCID: PMC9854481 DOI: 10.3390/antiox12010150] [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: 12/19/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/10/2023] Open
Abstract
Hypertension is associated with increased expression of kinin B1 receptors (B1R) and increased levels of pro-inflammatory cytokines within the neurons. We previously reported that angiotensin II (Ang II) upregulates B1R expression and can induce neuroinflammation and oxidative stress in primary hypothalamic neurons. However, the order in which B1R activation, neuroinflammation, and oxidative stress occur has not yet been studied. Using primary hypothalamic neurons from neonatal mice, we show that tumor necrosis factor (TNF), lipopolysaccharides (LPS), and hydrogen peroxide (H2O2) can upregulate B1R expression and increase oxidative stress. Furthermore, our study shows that B1R blockade with R715, a specific B1R antagonist, can attenuate these effects. To further confirm our findings, we used a deoxycorticosterone acetate (DOCA)-salt model of hypertension to show that oxidative stress is upregulated in the hypothalamic paraventricular nucleus (PVN) of the brain. Together, these data provide novel evidence that relationship between oxidative stress, neuroinflammation, and B1R upregulation in the brain is bidirectional, and that B1R antagonism may have beneficial effects on neuroinflammation and oxidative stress in various disease pathologies.
Collapse
|
30
|
Koziol SA, Oba PM, Soto-Diaz K, Steelman AJ, Suchodolski JS, Eckhardt ERM, Swanson KS. Effects of a Lactobacillus fermentation product on the fecal characteristics, fecal microbial populations, immune function, and stress markers of adult dogs. J Anim Sci 2023; 101:skad160. [PMID: 37208000 PMCID: PMC10237232 DOI: 10.1093/jas/skad160] [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: 03/15/2023] [Accepted: 05/15/2023] [Indexed: 05/21/2023] Open
Abstract
The objective of this study was to measure the effects of a Lactobacillus fermentation product (LBFP) on fecal characteristics and microbiota, blood biomarkers, immune function, and serum oxidative stress markers of adult dogs. Thirty adult beagle dogs [23 M, 7 F; mean age = 8.47 ± 2.65 yr old; mean BW = 15.43 ± 4.17 kg] were used in a completely randomized design study. All dogs were fed a basal diet to maintain BW for 5 wk, followed by baseline blood and fecal sample collections. Dogs remained on the same diet, but then were randomly assigned to a placebo (dextrose) or LBFP supplement (Limosilactobacillus fermentum and Lactobacillus delbrueckii). Both treatments were dosed at 4 mg/kg BW via gelatin capsule for 5 wk (n = 15/treatment). Fecal and blood samples were collected at that time. Change from baseline data were analyzed using the Mixed Models procedure of SAS 9.4, with P < 0.05 being significant and P < 0.10 being trends. Most circulating metabolites and immunoglobulins (Ig) were unaltered by treatment, but LBFP-supplemented dogs had lower changes in serum corticosteroid isoenzyme of alkaline phosphatase (P < 0.05), alanine aminotransferase (P < 0.10), and IgM (P < 0.10) than controls. The change in fecal scores tended to be lower (P = 0.068) in LBFP-supplemented dogs than controls, signifying firmer feces in LBFP-supplemented dogs. Regarding the fecal microbiota, alpha diversity indicators tended to be higher (P = 0.087) in LBFP-supplemented dogs than controls. One fecal bacterial phylum (Actinobacteriota) was altered by treatments, with its relative abundance tending to have a greater (P < 0.10) increase in controls than LBFP-supplemented dogs. Fifteen bacterial genera were altered (P < 0.05 or P < 0.10) by treatments, including relative abundances of fecal Peptoclostridium, Sarcina, and Faecalitalea that had a greater (P < 0.05) increase in controls than LBFP-supplemented dogs. In contrast, relative abundances of fecal Faecalibaculum, Bifidobacterium, and uncultured Butyricicoccaceae had a greater (P ≤ 0.05) increase in LBFP-supplemented dogs than controls. After week 5, dogs underwent transport stress (45-min vehicle ride) to assess oxidative stress markers. The change in serum superoxide dismutase after transport had a greater (P < 0.0001) increase in LBFP-supplemented dogs than controls. Our data suggest that LBFP may provide benefits to dogs by stabilizing stool quality, beneficially shifting fecal microbiota, and protecting against oxidative damage when subjected to stress.
Collapse
Affiliation(s)
- Samantha A Koziol
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Patricia M Oba
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Katiria Soto-Diaz
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Andrew J Steelman
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jan S Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA
| | | | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
31
|
Valverde-Pérez E, Olea E, Obeso A, Prieto-Lloret J, Rocher A, Gonzalez-Obeso E. Intermittent Hypoxia and Diet-Induced Obesity on the Intestinal Wall Morphology in a Murine Model of Sleep Apnea. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1427:89-97. [PMID: 37322339 DOI: 10.1007/978-3-031-32371-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
This work analyzes the impact of two conditions, intermittent hypoxia exposure and high-fat diet in rats as models of sleep apnea. We studied the autonomic activity and histological structure of the rat jejunum and whether the overlapping of both conditions, as often observed in patients, induces more deleterious effects on the intestinal barrier. We found alterations in jejunum wall histology, predominantly in HF rats, based on increased crypt depth and submucosal thickness, as well as decreased muscularis propria thickness. These alterations were maintained with the IH and HF overlap. An increase in the number and size of goblet cells in the villi and crypts and the infiltration of eosinophils and lymphocytes in the lamina propria suggest an inflammatory status, confirmed by the increase in plasma CRP levels in all experimental groups. Regarding the CAs analysis, IH, alone or combined with HF, causes a preferential accumulation of NE in the catecholaminergic nerve fibers of the jejunum. In contrast, serotonin increases in all three experimental conditions, with the highest level in the HF group. It remains to be elucidated whether the alterations found in the present work could affect the permeability of the intestinal barrier, promoting sleep apnea-induced morbidities.
Collapse
Affiliation(s)
- Esther Valverde-Pérez
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
| | - Elena Olea
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
- Departamento de Enfermería, Facultad de Enfermeria, Universidad de Valladolid, Valladolid, Spain
| | - Ana Obeso
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
| | - Jesús Prieto-Lloret
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
| | - Asunción Rocher
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain.
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain.
| | - Elvira Gonzalez-Obeso
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
- Servicio de Anatomía Patológica, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| |
Collapse
|
32
|
Chaudhari S, D'Souza BM, Morales JY, Young-Stubbs CM, Shimoura CG, Ma R, Mathis KW. Renal TLR-7/TNF-α pathway as a potential female-specific mechanism in the pathogenesis of autoimmune-induced hypertension. Am J Physiol Heart Circ Physiol 2022; 323:H1331-H1342. [PMID: 36367687 PMCID: PMC9744658 DOI: 10.1152/ajpheart.00286.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/04/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
Hypertension is prevalent in patients with systemic lupus erythematosus (SLE). The goal of the current study is to track the pathogenesis of hypertension and renal injury in SLE, identify contributory mechanisms, and highlight differences in disease development among sexes. Mean arterial pressure was measured in conscious male and female SLE (NZBWF1) and control (NZW) mice at 34-35 wk of age using indwelling arterial catheters. Measures of renal injury, renal inflammation, and renal hemodynamics were used to monitor the potential contributors to latent sex differences. Both male and female SLE mice were hypertensive at 35 wk of age, and the hypertension was linked to renal injury in females, but not in males. A known contributor of renal pathology in SLE, Toll-like receptor (TLR)-7, and its downstream effector, the proinflammatory cytokine tumor necrosis factor (TNF)-α, were lower in male SLE mice than in females. Male SLE mice also had higher glomerular filtration rate (GFR) and lower renal vascular resistance (RVR) than females. Our data suggest that although hypertension in female SLE mice is associated with renal mechanisms, hypertension in male SLE mice may develop independent of renal changes. Future studies will continue to dissect sex-specific factors that should be considered when treating patients with hypertension with underlying chronic inflammation and/or autoimmunity.NEW & NOTEWORTHY There is a high prevalence of hypertension in male and female SLE; however, male SLE mice are hypertensive without renal involvement. The development of hypertension in female SLE mice is renocentric and strongly associated with injurious renal mechanisms like the TLR-7→TNF-α pathway. This clear difference in the pathogenesis among the sexes could have a significant impact on how we treat patients with hypertension with underlying chronic autoimmune/inflammatory diseases.
Collapse
Affiliation(s)
- Sarika Chaudhari
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Bradley M D'Souza
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Jessica Y Morales
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Cassandra M Young-Stubbs
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Caroline G Shimoura
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Keisa W Mathis
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
33
|
Geerling E, Hameed M, Weger-Lucarelli J, Pinto AK. Metabolic syndrome and aberrant immune responses to viral infection and vaccination: Insights from small animal models. Front Immunol 2022; 13:1015563. [PMID: 36532060 PMCID: PMC9747772 DOI: 10.3389/fimmu.2022.1015563] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
This review outlines the propensity for metabolic syndrome (MetS) to induce elevated disease severity, higher mortality rates post-infection, and poor vaccination outcomes for viral pathogens. MetS is a cluster of conditions including high blood glucose, an increase in circulating low-density lipoproteins and triglycerides, abdominal obesity, and elevated blood pressure which often overlap in their occurrence. MetS diagnoses are on the rise, as reported cases have increased by greater than 35% since 1988, resulting in one-third of United States adults currently diagnosed as MetS patients. In the aftermath of the 2009 H1N1 pandemic, a link between MetS and disease severity was established. Since then, numerous studies have been conducted to illuminate the impact of MetS on enhancing virally induced morbidity and dysregulation of the host immune response. These correlative studies have emphasized the need for elucidating the mechanisms by which these alterations occur, and animal studies conducted as early as the 1940s have linked the conditions associated with MetS with enhanced viral disease severity and poor vaccine outcomes. In this review, we provide an overview of the importance of considering overall metabolic health in terms of cholesterolemia, glycemia, triglyceridemia, insulin and other metabolic molecules, along with blood pressure levels and obesity when studying the impact of metabolism-related malignancies on immune function. We highlight the novel insights that small animal models have provided for MetS-associated immune dysfunction following viral infection. Such animal models of aberrant metabolism have paved the way for our current understanding of MetS and its impact on viral disease severity, dysregulated immune responses to viral pathogens, poor vaccination outcomes, and contributions to the emergence of viral variants.
Collapse
Affiliation(s)
- Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Muddassar Hameed
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - James Weger-Lucarelli
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
34
|
Soh HS, Cho B. Long COVID-19 and Health-Related Quality of Life of Mild Cases in Korea: 3-Months Follow-up of a Single Community Treatment Center. J Korean Med Sci 2022; 37:e326. [PMID: 36631024 PMCID: PMC9705205 DOI: 10.3346/jkms.2022.37.e326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (coronavirus disease 2019 [COVID-19]) survivors mount as the pandemic continues. To date, studies on persistent symptoms and their effects on health-related quality of life (HRQoL) in mild COVID-19 cases have been limited. METHODS A prospective online survey was conducted in COVID-19 patients who were admitted to Seongnam Community Treatment Center, an isolation center in South Korea, from November 23rd 2021 to January 2nd 2022. Patients above the age of 19 with no or mild symptoms were included in the study. Total of 147 patients returned to the follow-up survey 3 months after discharge. Baseline demographics, clinical characteristics, symptoms, and EuroQol-5 dimensions-5 levels (EQ-5D-5L) measures were investigated. RESULTS The median (interquartile range [IQR]) interval period between the initial and follow-up survey was 96.0 (93.0-98.0) days. The median (IQR) age of participants was 51.0 (43.0-61.0). During isolation, 131 (89.1%) patients manifested symptoms. On follow-up, 82 (55.8%) participants remained symptomatic. Common symptoms were constitutional (fatigue, myalgia), neurological (memory impairment, hyposmia, hypogeusia, dizziness), and neuropsychiatric (anxiety) symptoms. Participants with remaining neuropsychiatric symptoms reported the lowest EQ-5D-5L index values. Factors associated with persistent symptoms and diminished HRQoL were identified as female sex, metabolic disease, and anxiety during acute COVID-19 phase. CONCLUSIONS Cardiopulmonary symptoms improved over time but constitutional, neurological, neuropsychiatric symptoms remained. Patients with neuropsychiatric symptoms (anxiety and insomnia) reported the worst HRQoL. Female sex, metabolic disease, and anxiety during the acute COVID-19 phase were associated with long COVID. Observations of long-term symptoms of COVID-19 with decline in HRQoL and integrated research in COVID-19 survivors are warranted.
Collapse
Affiliation(s)
- Hi Sun Soh
- Department of Family Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Clinical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - BeLong Cho
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
- Institute on Aging, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
35
|
Angiotensin II Modulates Calcium/Phosphate Excretion in Experimental Model of Hypertension: Focus on Bone. Biomedicines 2022; 10:biomedicines10112928. [PMID: 36428495 PMCID: PMC9687632 DOI: 10.3390/biomedicines10112928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
A link between hypertension and long-term bone health has been suggested. The aim of this study was to investigate the effects of chronic angiotensin II administration on urinary calcium/phosphate excretion, bone mineral density, bone remodeling and osteoblast population in a well-established experimental model of hypertension, in the absence of possible confounding factors that could affect bone metabolism. Male Sprague-Dawley rats, divided in the following groups: (a) Angiotensin II (Ang II, 200 ng/kg/min, osmotic minipumps, sub cutis, n = 8); (b) Ang II+losartan (Los, 50 mg/kg/day, per os, n = 6); (c) control group (physiological saline, sub cutis, n = 9); and (d) control+losartan (n = 6) were treated for four weeks. During the experimental period, 24-hour diuresis, urinary calcium, phosphate and sodium excretion were measured prior to the treatment, at two weeks of treatment, and at the end of the treatment. Systolic blood pressure was measured by plethysmography technique (tail cuff method). At the end of the experimental protocol, the rats were euthanized and peripheral quantitative computed tomography at the proximal metaphysis and at the diaphysis of the tibiae and quantitative bone histomorphometry on distal femora were performed. Angiotensin II-dependent hypertension is associated with increased calcium and phosphate excretion. AT1 receptor blockade prevented the increase of blood pressure and phosphate excretion but did not affect the increase of calcium excretion. These changes took place without significantly affecting bone density, bone histology or osteoblast population. In conclusion, in our experimental conditions, angiotensin II-dependent hypertension gave rise to an increased urinary excretion of calcium and phosphate without affecting bone density.
Collapse
|
36
|
Xu J, Moore BN, Pluznick JL. Short-Chain Fatty Acid Receptors and Blood Pressure Regulation: Council on Hypertension Mid-Career Award for Research Excellence 2021. Hypertension 2022; 79:2127-2137. [PMID: 35912645 PMCID: PMC9458621 DOI: 10.1161/hypertensionaha.122.18558] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The gut microbiome influences host physiology and pathophysiology through several pathways, one of which is microbial production of chemical metabolites which interact with host signaling pathways. Short-chain fatty acids (SCFAs) are a class of gut microbial metabolites known to activate multiple signaling pathways in the host. Growing evidence indicates that the gut microbiome is linked to blood pressure, that SCFAs modulate blood pressure regulation, and that delivery of exogenous SCFAs lowers blood pressure. Given that hypertension is a key risk factor for cardiovascular disease, the examination of novel contributors to blood pressure regulation has the potential to lead to novel approaches or treatments. Thus, this review will discuss SCFAs with a focus on their host G protein-coupled receptors including GPR41 (G protein-coupled receptor 41), GPR43, and GPR109A, as well as OLFR78 (olfactory receptor 78) and OLFR558. This includes a discussion of the ligand profiles, G protein coupling, and tissue distribution of each receptor. We will also review phenotypes relevant to blood pressure regulation which have been reported to date for Gpr41, Gpr43, Gpr109a, and Olfr78 knockout mice. In addition, we will consider how SCFA signaling influences physiology at baseline, and, how SCFA signaling may contribute to blood pressure regulation in settings of hypertension. In sum, this review will integrate current knowledge regarding how SCFAs and their receptors regulate blood pressure.
Collapse
Affiliation(s)
- Jiaojiao Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Brittni N. Moore
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jennifer L. Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
37
|
Ng WY, Atan R, Mohd Yunos N, bin Md Kamal AH, Roslan MH, Quah KY, Teh KX, Zaid M, Kassim M, Mariapun J, Ngim CF, Dhanoa A, Yeo TW. A double whammy: The association between comorbidities and severe dengue among adult patients—A matched case-control study. PLoS One 2022; 17:e0273071. [PMID: 36126060 PMCID: PMC9488767 DOI: 10.1371/journal.pone.0273071] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
Background Dengue infection is the most prevalent mosquito-borne viral infection globally. Concurrently, there has also been an upsurge of non-communicable comorbidities. We aimed to investigate the association between these comorbidities and the development of severe dengue. Methods We performed a retrospective, case-control study involving 117 cases with severe dengue and 351 controls with non-severe dengue; matched according to gender, age (+/- 5 years old), and admission date (+/- 2 weeks). We analyzed the data using conditional odds ratio (cOR) and adjusted conditional odds ratio (AcOR) using univariate and multivariable conditional logistic regression respectively. Results Six main comorbidities namely obesity, diabetes mellitus, hypertension, hyperlipidemia, chronic pulmonary disease, and ischemic heart disease were observed among cases and controls. Multivariable conditional logistic regression model found only hypertension to be independently associated with the development of severe dengue (ACOR 2.46; 95% CI:1.09–5.53). Among symptoms at presentation, lethargy, vomiting, bleeding manifestations, and abdominal pain were associated with increased odds of severe dengue, although the associations were not statistically significant. Headache (ACOR: 0:32; 95% CI: 0.21–0.51) and skin rash (ACOR: 0.42; 95% CI: 0.22–0.81) were associated with significantly lower odds of severe dengue. Severe dengue patients were also found to have significantly higher white cell count, urea, creatinine, alanine aminotransferase, aspartate aminotransferase, creatine kinase, and lactate dehydrogenase on admission, while platelet and albumin were significantly lower compared to non-severe dengue patients. Conclusions Our study found a significant association between hypertension and the development of severe dengue in adult patients. For clinical practice, this finding suggests that dengue patients with underlying hypertension warrant closer clinical monitoring for deterioration. The association between significant derangement in various laboratory parameters and severe dengue as shown in this study is in keeping with previous reports. While further substantiation by larger prospective studies will be desirable, this association may serve to inform the dengue triaging process.
Collapse
Affiliation(s)
- Wei Yao Ng
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
- Mackay Base Hospital, Mackay Hospital and Health Services, West Mackay, Queensland, Australia
| | - Rafidah Atan
- Department of Anesthesiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Nor’azim Mohd Yunos
- Department of Anesthesiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Adam Harrish bin Md Kamal
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- * E-mail:
| | - Mohd Hariz Roslan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Kai Yuan Quah
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
- Mackay Base Hospital, Mackay Hospital and Health Services, West Mackay, Queensland, Australia
| | - Kai Xuan Teh
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Masliza Zaid
- Department of Medicine, Hospital Sultanah Aminah, Johor Bahru, Johor, Malaysia
| | - Mahazir Kassim
- Department of Anesthesiology and Intensive Care, Hospital Sultanah Aminah, Johor Bahru, Johor, Malaysia
| | - Jeevitha Mariapun
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Chin Fang Ngim
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Amreeta Dhanoa
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Tsin Wen Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University and Imperial College, Singapore, Singapore
| |
Collapse
|
38
|
Barsukov AV, Korovin AE, Churilov LP, Borisova EV, Tovpeko DV. Heart Dysfunction in Essential Hypertension Depends on Systemic Proinflammatory Influences: A Retrospective Clinical Pathophysiological Study. PATHOPHYSIOLOGY 2022; 29:453-468. [PMID: 35997392 PMCID: PMC9396991 DOI: 10.3390/pathophysiology29030036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 11/28/2022] Open
Abstract
Low-intensity systemic inflammation is an important element of heart failure pathogenesis. The aim of this study is to assess proinflammatory status serum indicators (C-reactive protein (CRP), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6)) in middle-aged males (M) and females (F) with essential hypertension (HTN) depending on left ventricular (LV) diastolic dysfunction (LVDD). The main group comprised 55 M and 49 F with the first- to second-severity grade HTN with mild heart failure and a preserved LV ejection fraction ≥50%. Patients had sinus rhythm, first or second-severity degree LVDD, LV hypertrophy, left atrium dilatation, and NT-proBNP > 125 pg/mL. Comparison group: 30 hypertensives without cardiac dysfunction; control group: 31 normotensives. Quantitative features were compared using the Mann−Whitney test, median χ2, ANOVA module. Spearman’s rank correlation coefficients were determined to identify the relationship between the proinflammatory pattern and exercise tolerance. Hypertensive M had markedly higher CRP, TNF-α, and IL-6 levels compared to F. All mean values corresponded to reference range. In patients with second-degree LVDD, CRP, TNF-α, and IL-6 levels were significantly greater than in subjects with first-degree LVDD (both within M and within F samples). Significant negative associations between CRP, IL-6, and TNF-α levels and the 6 min walk test existed in hypertensive M and F. The study demonstrated a close relationship between the proinflammatory pattern and LVDD and exercise tolerance indicators, regardless of the hypertensive patient’s sex.
Collapse
Affiliation(s)
| | - Alexander E. Korovin
- Laboratory of the Microangiopathic Mechanisms of Atherogenesis, Department of Pathology, Saint Petersburg State University, 199034 St. Petersburg, Russia
- S.M. Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - Leonid P. Churilov
- Laboratory of the Microangiopathic Mechanisms of Atherogenesis, Department of Pathology, Saint Petersburg State University, 199034 St. Petersburg, Russia
- Saint Petersburg Research Institute of Phthisiopulmonology, 194064 St. Petersburg, Russia
| | | | | |
Collapse
|
39
|
Liu Q, Chi S, Dmytruk K, Dmytruk O, Tan S. Coronaviral Infection and Interferon Response: The Virus-Host Arms Race and COVID-19. Viruses 2022; 14:v14071349. [PMID: 35891331 PMCID: PMC9325157 DOI: 10.3390/v14071349] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 02/07/2023] Open
Abstract
The recent pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in unprecedented morbidity and mortality worldwide. The host cells use a number of pattern recognition receptors (PRRs) for early detection of coronavirus infection, and timely interferon secretion is highly effective against SARS-CoV-2 infection. However, the virus has developed many strategies to delay interferon secretion and disarm cellular defense by intervening in interferon-associated signaling pathways on multiple levels. As a result, some COVID-19 patients suffered dramatic susceptibility to SARS-CoV-2 infection, while another part of the population showed only mild or no symptoms. One hypothesis suggests that functional differences in innate immune integrity could be the key to such variability. This review tries to decipher possible interactions between SARS-CoV-2 proteins and human antiviral interferon sensors. We found that SARS-CoV-2 actively interacts with PRR sensors and antiviral pathways by avoiding interferon suppression, which could result in severe COVID-19 pathogenesis. Finally, we summarize data on available antiviral pharmaceutical options that have shown potential to reduce COVID-19 morbidity and mortality in recent clinical trials.
Collapse
Affiliation(s)
- Qi Liu
- Department of Immunology, School of Basic Medicine, Chongqing Medical University, Chongqing 400010, China;
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: (Q.L.); (S.T.)
| | - Sensen Chi
- Department of Immunology, School of Basic Medicine, Chongqing Medical University, Chongqing 400010, China;
| | - Kostyantyn Dmytruk
- Department of Molecular Genetics and Biotechnology, Institute of Cell Biology, National Academy of Sciences of Ukraine, 79005 Lviv, Ukraine; (K.D.); (O.D.)
| | - Olena Dmytruk
- Department of Molecular Genetics and Biotechnology, Institute of Cell Biology, National Academy of Sciences of Ukraine, 79005 Lviv, Ukraine; (K.D.); (O.D.)
- Institute of Biology and Biotechnology, University of Rzeszow, 35-601 Rzeszow, Poland
| | - Shuai Tan
- Department of Immunology, School of Basic Medicine, Chongqing Medical University, Chongqing 400010, China;
- Correspondence: (Q.L.); (S.T.)
| |
Collapse
|
40
|
Ebinger JE, Joung S, Liu Y, Wu M, Weber B, Claggett B, Botting PG, Sun N, Driver M, Kao YH, Khuu B, Wynter T, Nguyen TT, Alotaibi M, Prostko JC, Frias EC, Stewart JL, Goodridge HS, Chen P, Jordan SC, Jain M, Sharma S, Fert-Bober J, Van Eyk JE, Minissian MB, Arditi M, Melmed GY, Braun JG, McGovern DPB, Cheng S, Sobhani K. Demographic and clinical characteristics associated with variations in antibody response to BNT162b2 COVID-19 vaccination among healthcare workers at an academic medical centre: a longitudinal cohort analysis. BMJ Open 2022; 12:e059994. [PMID: 35613792 PMCID: PMC9130668 DOI: 10.1136/bmjopen-2021-059994] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/11/2022] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES We sought to understand the demographic and clinical factors associated with variations in longitudinal antibody response following completion of two-dose regiment of BNT162b2 vaccination. DESIGN This study is a 10-month longitudinal cohort study of healthcare workers and serially measured anti-spike protein IgG (IgG-S) antibody levels using mixed linear models to examine their associations with participant characteristics. SETTING A large, multisite academic medical centre in Southern California, USA. PARTICIPANTS A total of 843 healthcare workers met inclusion criteria including completion of an initial two-dose course of BNT162b2 vaccination, complete clinical history and at least two blood samples for analysis. Patients had an average age of 45±13 years, were 70% female and 7% with prior SARS-CoV-2 infection. RESULTS Vaccine-induced IgG-S levels remained in the positive range for 99.6% of individuals up to 10 months after initial two-dose vaccination. Prior SARS-CoV-2 infection was the primary correlate of sustained higher postvaccination IgG-S levels (partial R2=0.133), with a 1.74±0.11 SD higher IgG-S response (p<0.001). Female sex (beta 0.27±0.06, p<0.001), younger age (0.01±0.00, p<0.001) and absence of hypertension (0.17±0.08, p=0.003) were also associated with persistently higher IgG-S responses. Notably, prior SARS-CoV-2 infection augmented the associations of sex (-0.42 for male sex, p=0.08) and modified the associations of hypertension (1.17, p=0.001), such that infection-naïve individuals with hypertension had persistently lower IgG-S levels whereas prior infected individuals with hypertension exhibited higher IgG-S levels that remained augmented over time. CONCLUSIONS While the IgG-S antibody response remains in the positive range for up to 10 months following initial mRNA vaccination in most adults, determinants of sustained higher antibody levels include prior SARS-CoV-2 infection, female sex, younger age and absence of hypertension. Certain determinants of the longitudinal antibody response appear significantly modified by prior infection status. These findings offer insights regarding factors that may influence the 'hybrid' immunity conferred by natural infection combined with vaccination.
Collapse
Affiliation(s)
- Joseph E Ebinger
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sandy Joung
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yunxian Liu
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Min Wu
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Brittany Weber
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Brian Claggett
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Patrick G Botting
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nancy Sun
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Matthew Driver
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yu Hung Kao
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Briana Khuu
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Timothy Wynter
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Trevor-Trung Nguyen
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mona Alotaibi
- Division of Pulmonary and Critical Care Medicine, University of California San Diego, San Diego, California, USA
| | - John C Prostko
- Applied Research and Technology, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Edwin C Frias
- Applied Research and Technology, Abbott Laboratories, Abbott Park, Illinois, USA
| | - James L Stewart
- Applied Research and Technology, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Helen S Goodridge
- Department of Biomedical Sciences, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Peter Chen
- Department of Biomedical Sciences, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Stanley C Jordan
- Transplant Immunology Laboratory and Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mohit Jain
- Department of Medicine, School of Medicine, University of California, San Diego, San Diego, California, USA
| | - Sonia Sharma
- La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Justyna Fert-Bober
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jennifer E Van Eyk
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Advanced Clinical Biosystems Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Margo B Minissian
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Brawerman Nursing Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Moshe Arditi
- Smidt Heart Institute; Department of Pediatrics, Division of Infectious Diseases and Immunology; Infectious and Immunologic Diseases Research Center (IIDRC); Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Gil Y Melmed
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jonathan G Braun
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dermot P B McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kimia Sobhani
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
41
|
Notarte KI, Ver AT, Velasco JV, Pastrana A, Catahay JA, Salvagno GL, Yap EPH, Martinez-Sobrido L, B Torrelles J, Lippi G, Henry BM. Effects of age, sex, serostatus, and underlying comorbidities on humoral response post-SARS-CoV-2 Pfizer-BioNTech mRNA vaccination: a systematic review. Crit Rev Clin Lab Sci 2022; 59:373-390. [PMID: 35220860 PMCID: PMC8935447 DOI: 10.1080/10408363.2022.2038539] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the advent of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic, several vaccines have been developed to mitigate its spread and prevent adverse consequences of the Coronavirus Disease 2019 (COVID-19). The mRNA technology is an unprecedented vaccine, usually given in two doses to prevent SARS-CoV-2 infections. Despite effectiveness and safety, inter-individual immune response heterogeneity has been observed in recipients of mRNA-based vaccines. As a novel disease, the specific immune response mechanism responsible for warding off COVID-19 remains unclear at this point. However, significant evidence suggests that humoral response plays a crucial role in affording immunoprotection and preventing debilitating sequelae from COVID-19. As such, this paper focused on the possible effects of age, sex, serostatus, and comorbidities on humoral response (i.e. total antibodies, IgG, and/or IgA) of different populations post-mRNA-based Pfizer-BioNTech vaccination. A systematic search of literature was performed through PubMed, Cochrane CENTRAL, Google Scholar, Science Direct, medRxiv, and Research Square. Studies were included if they reported humoral response to COVID-19 mRNA vaccines. A total of 32 studies were identified and reviewed, and the percent differences of means of reported antibody levels were calculated for comparison. Findings revealed that older individuals, male sex, seronegativity, and those with more comorbidities mounted less humoral immune response. Given these findings, several recommendations were proposed regarding the current vaccination practices. These include giving additional doses of vaccination for immunocompromised and elderly populations. Another recommendation is conducting clinical trials in giving a combined scheme of mRNA vaccines, protein vaccines, and vector-based vaccines.
Collapse
Affiliation(s)
- Kin Israel Notarte
- Faculty of Medicine and Surgery, University of Santo Tomas, Manila, Philippines
| | | | | | - Adriel Pastrana
- Faculty of Medicine and Surgery, University of Santo Tomas, Manila, Philippines
| | | | - Gian Luca Salvagno
- Service of Laboratory Medicine, Pederzoli Hospital, Peschiera del Garda, Italy.,Section of Clinical Biochemistry, University of Verona, Verona, Italy
| | - Eric Peng Huat Yap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Luis Martinez-Sobrido
- Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Jordi B Torrelles
- Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| | - Brandon Michael Henry
- Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA.,Division of Nephrology and Hypertension, Clinical Laboratory, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
42
|
Dynamics of Soluble Factors and Double-Negative T Cells Associated with Response to Renal Denervation in Resistant Hypertension Patients. J Pers Med 2022; 12:jpm12030343. [PMID: 35330342 PMCID: PMC8950746 DOI: 10.3390/jpm12030343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 12/10/2022] Open
Abstract
The role of the immune system, and hence inflammation, in the pathophysiology of hypertensive patients is not clear. Until now, most clinical and biochemical parameters have failed to predict a positive response to renal denervation (RDN). Our aim was to evaluate the immune response in a cohort of patients treated by RDN, through the analysis of cytokine, chemokine, and growth factor behavior. A population of 21 resistant hypertension patients, treated by RDN, was evaluated at six months and one year. Response was defined as a drop of ≥5 mmHg in ambulatory blood pressure monitoring. Sixty-seven percent and 81% of patients clinically responded after six months and one year, respectively. There were no complications or safety issues. Plasmatic levels of 45 cytokine, chemokine, and growth factors were quantified at four different times, pre- and post-procedure. Baseline characteristics were similar between groups, except that active smoking was more frequent in non-responders at one year. Regulated on activation, normal T cell expressed, and secreted (RANTES/CCL5) levels were significantly lower in responders, both at baseline and at 30 days (p = 0.037), and a level ≤15,496 pg/mL was the optimal cutoff, for prediction of a response. IL-15, IL-17A, IL-27, and leukemia inhibitory factor varied significantly in time, with an acute rise being observed 24 h after RDN. Our group has previously showed that HLA-DR+ double-negative (DN) T cells were significantly lower in responders. There was a positive correlation between IL-13, -27, and -4, and DN T cells, and a negative correlation between the latter and SDF-1α and TNF-α, at baseline. Low plasmatic levels of the chemokine RANTES/CCL5 was the most significant result associated with RDN response and may help to identify the best candidates among patients with true resistant hypertension. Pro-inflammatory cytokines correlated negatively with DN T cells in responders, a finding compatible with an enhanced inflammatory milieu present in this extremely high cardiovascular risk cohort.
Collapse
|
43
|
Pramudita A, Rosidah S, Yudia N, Simatupang J, Sigit WP, Novariani R, Myriarda P, Siswanto BB. Cardiometabolic Morbidity and Other Prognostic Factors for Mortality in Adult Hospitalized COVID-19 Patients in North Jakarta, Indonesia. Glob Heart 2022; 17:9. [PMID: 35342692 PMCID: PMC8855735 DOI: 10.5334/gh.1019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 01/24/2022] [Indexed: 01/08/2023] Open
Abstract
Background Although there have been several studies investigating prognostic factors for mortality in COVID-19, there have been lack of studies in low- and middle-income countries, including Indonesia. To date, the country has the highest mortality rate among Asian countries. Objective We sought to identify the prognostic factors of mortality in hospitalized patients with COVID-19 in Jakarta. Methods In this retrospective cohort study, we included all adult inpatients (≥18 years old) with confirmed COVID-19 from Koja General Hospital (North Jakarta, Indonesia) who had been hospitalized between March 20th and July 31st, 2020. Demographic, clinical, laboratory, and radiology data were extracted from the medical records and compared between survivors and non-survivors. Univariate and multivariate logistic regression analysis were used to explore the prognostic factors associated with in-hospital death. Results Two hundred forty-three patients were included in the study, of whom 32 died. Comorbid of hypertension (OR 3.59; 95% CI 1.12-11.48; p = 0.031), obesity (OR 6.34; 95% CI 1.68-23.98; p = 0.007), immediate need of HFNC and/or IMV (OR 64.93; 95% CI 11.08-380.61; p < 0.001), abnormal RDW (OR 3.68; 95% CI 1.09-12.34; p = 0.035), ALC < 1,000/µL (OR 3.51; 95% CI 1.08-11.44; p = 0.038), D-dimer > 500 ng/mL (OR 9.36; 95% CI 1.53-57.12; p = 0.015) on admission, as well as chloroquine treatment (OR 3.61; 95% CI 1.09-11.99; p = 0.036) were associated with greater risk of overall mortality in COVID-19 patients. The likelihood of mortality increased with increasing number of prognostic factors. Conclusion The potential prognostic factors of hypertension, obesity, immediate need of HFNC and/or IMV, abnormal RDW, ALC < 1,000/µL, D-dimer > 500 ng/mL, and chloroquine treatment could help clinicians to identify COVID-19 patients with poor prognosis at an early stage.
Collapse
Affiliation(s)
- Arvin Pramudita
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia, National Cardiovascular Center Harapan Kita, Jakarta, ID
- Koja General Hospital, Jakarta, ID
| | | | | | | | | | | | | | - Bambang Budi Siswanto
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia, National Cardiovascular Center Harapan Kita, Jakarta, ID
| |
Collapse
|
44
|
Neely OC, Domingos AI, Paterson DJ. Macrophages Can Drive Sympathetic Excitability in the Early Stages of Hypertension. Front Cardiovasc Med 2022; 8:807904. [PMID: 35155614 PMCID: PMC8828732 DOI: 10.3389/fcvm.2021.807904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/21/2021] [Indexed: 12/31/2022] Open
Abstract
Hypertension is a major health burden worldwide with many cases resistant to current treatments. Hyperactivity of the sympathetic nervous contributes to the etiology and progression of the disease, where emerging evidence suggests that inflammation may underpin the development of sympathetic dysautonomia. This study examined whether macrophages could drive the sympathetic phenotype in Spontaneously Hypertensive Rats (SHR) before animals develop high pressure. Stellate neurons from wild-type control Wistar rats and SHRs were co-cultured with blood leukocytes from their own strain, and also crossed cultured between strains. The calcium transient response to nicotinic stimulation was recorded using Fura-2 calcium imaging, where SHR neurons had a greater calcium transient compared with Wistar neurons. However, when co-cultured with leukocytes, Wistar neurons began to phenocopy the SHR sympathetic hyperactivity, while the SHR neurons themselves were unaltered. Resident leukocyte populations of the SHR and Wistar stellate ganglia were then compared using flow cytometry, where there was a shift in monocyte-macrophage subset proportions. While classical monocyte-macrophages were predominant in the Wistar, there were relatively more of the non-classical subset in the SHR, which have been implicated in pro-inflammatory roles in a number of diseases. When bone marrow-derived macrophages (BMDMs) were co-cultured with stellate neurons, they made Wistar neurons recapitulate the SHR nicotinic stimulated calcium transient. Wistar BMDMs however, had no effect on SHR neurons, even though SHR BMDMs increased SHR neuron responsiveness further above their hyper-responsive state. Taken together, these findings show that macrophages can be potent enhancers of sympathetic neuronal calcium responsiveness, and thus could conceivably play a role in peripheral sympathetic hyperactivity observed in the early stages of hypertension.
Collapse
Affiliation(s)
- Oliver C Neely
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Ana I Domingos
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - David J Paterson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Canoy D, Harvey NC, Prieto-Alhambra D, Cooper C, Meyer HE, Åsvold BO, Nazarzadeh M, Rahimi K. Elevated blood pressure, antihypertensive medications and bone health in the population: revisiting old hypotheses and exploring future research directions. Osteoporos Int 2022; 33:315-326. [PMID: 34642814 PMCID: PMC8813726 DOI: 10.1007/s00198-021-06190-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/01/2021] [Indexed: 11/17/2022]
Abstract
Blood pressure and bone metabolism appear to share commonalities in their physiologic regulation. Specific antihypertensive drug classes may also influence bone mineral density. However, current evidence from existing observational studies and randomised trials is insufficient to establish causal associations for blood pressure and use of blood pressure-lowering drugs with bone health outcomes, particularly with the risks of osteoporosis and fractures. The availability and access to relevant large-scale biomedical data sources as well as developments in study designs and analytical approaches provide opportunities to examine the nature of the association between blood pressure and bone health more reliably and in greater detail than has ever been possible. It is unlikely that a single source of data or study design can provide a definitive answer. However, with appropriate considerations of the strengths and limitations of the different data sources and analytical techniques, we should be able to advance our understanding of the role of raised blood pressure and its drug treatment on the risks of low bone mineral density and fractures. As elevated blood pressure is highly prevalent and blood pressure-lowering drugs are widely prescribed, even small effects of these exposures on bone health outcomes could be important at a population level.
Collapse
Affiliation(s)
- D Canoy
- Deep Medicine, Nuffield Department of Women's and Reproductive Health, University of Oxford, Hayes House 1F, George St., Oxford, OX1 2BQ, UK.
- NIHR Oxford Biomedical Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - N C Harvey
- MRC Life Course Epidemiology Unit, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - D Prieto-Alhambra
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - C Cooper
- NIHR Oxford Biomedical Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- MRC Life Course Epidemiology Unit, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - H E Meyer
- Department of Community Medicine and Global Health, Faculty of Medicine, Oslo, Norway
- Norwegian Institute of Public Health, Oslo, Norway
| | - B O Åsvold
- Department of Endocrinology, Clinic of Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
| | - M Nazarzadeh
- Deep Medicine, Nuffield Department of Women's and Reproductive Health, University of Oxford, Hayes House 1F, George St., Oxford, OX1 2BQ, UK
| | - K Rahimi
- Deep Medicine, Nuffield Department of Women's and Reproductive Health, University of Oxford, Hayes House 1F, George St., Oxford, OX1 2BQ, UK
- NIHR Oxford Biomedical Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
46
|
Davidson JA, Banerjee A, Smeeth L, McDonald HI, Grint D, Herrett E, Forbes H, Pebody R, Warren-Gash C. Risk of acute respiratory infection and acute cardiovascular events following acute respiratory infection among adults with increased cardiovascular risk in England between 2008 and 2018: a retrospective, population-based cohort study. Lancet Digit Health 2021; 3:e773-e783. [PMID: 34823706 PMCID: PMC8628002 DOI: 10.1016/s2589-7500(21)00203-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/16/2021] [Accepted: 08/07/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Although acute respiratory infections can lead to cardiovascular complications, the effect of underlying cardiovascular risk on the incidence of acute respiratory infections and cardiovascular complications following acute respiratory infection in individuals without established cardiovascular disease is unknown. We aimed to investigate whether cardiovascular risk is associated with increased risk of acute respiratory infection and acute cardiovascular events after acute respiratory infection using 10 years of linked electronic health record (EHR) data in England. METHODS In this retrospective, population-based cohort study we used EHRs from primary care providers registered on the Clinical Practice Research Datalink (CPRD) GOLD and Aurum databases in England. Eligible individuals were aged 40-64 years, did not have established cardiovascular disease or a chronic health condition that would make them eligible for influenza vaccination, were registered at a general practice contributing to the CPRD, and had linked Hospital Episode Statistics Admitted Patient Care data in England from Sept 1, 2008, to Aug 31, 2018. We classified cardiovascular risk on the basis of diagnosed hypertension and overall predicted cardiovascular risk, estimated by use of the QRISK2 risk-prediction tool (comparing a score of ≥10% [increased risk] with a score of <10% [low risk]). Using multivariable Poisson regression models, we calculated incidence rate ratios (IRRs) for systemic acute respiratory infection. Among individuals who had an acute respiratory infection, we used multivariable Cox regression to calculate hazard ratios (HRs) for the risk of acute cardiovascular events within 1 year of infection. FINDINGS We identified 6 075 321 individuals aged 40-64 years with data in the CPRD and linked data in the Hospital Episode Statistics Admitted Patient Care database between Sept 1, 2008, and Aug 31, 2018. Of these individuals, 4 212 930 (including 526 480 [12·5%] with hypertension and 607 087 [14·4%] with a QRISK2 score of ≥10%) were included in the assessment of the incidence of acute respiratory infection. After adjusting for confounders (age, sex, ethnicity, socioeconomic status, body-mass index, alcohol consumption, smoking status, and consultation frequency in the hypertension analysis; and alcohol consumption and consultation frequency in the QRISK2 analysis), the incidence of acute respiratory infection was higher in individuals with hypertension than those without (IRR 1·04 [95% CI 1·03-1·05]) and higher in those with a QRISK2 score of 10% or higher than in those with a QRISK2 score of less than 10% (1·39 [1·37-1·40]). Of the 442 408 individuals who had an acute respiratory infection, 4196 (0·9%) had an acute cardiovascular event within 1 year of infection. After adjustment (for age, sex, ethnicity, socioeconomic status, body-mass index, alcohol consumption, and smoking status in the hypertension analysis; and for alcohol consumption in the QRISK2 analysis), hypertension (HR 1·98 [95% CI 1·83-2·15]) and a QRISK2 score of 10% or higher (3·65 [3·42-3·89]) were associated with a substantially increased risk of acute cardiovascular events after acute respiratory infection. INTERPRETATION People with increased cardiovascular risk but without diagnosed cardiovascular disease, measured by diagnosed hypertension or overall predicted cardiovascular risk, could benefit from influenza and pneumococcal vaccine prioritisation to reduce their risk of both acute respiratory infection and cardiovascular complications following an acute respiratory infection. FUNDING British Heart Foundation and the Wellcome Trust.
Collapse
Affiliation(s)
- Jennifer A Davidson
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK.
| | - Amitava Banerjee
- Institute of Health Informatics, University College London, London, UK
| | - Liam Smeeth
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; National Institute for Health Research Health Protection Research Unit in Immunisation, London School of Hygiene & Tropical Medicine, London, UK
| | - Helen I McDonald
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; National Institute for Health Research Health Protection Research Unit in Immunisation, London School of Hygiene & Tropical Medicine, London, UK
| | - Daniel Grint
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Emily Herrett
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Harriet Forbes
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Richard Pebody
- National Infection Service, Public Health England, London, UK
| | - Charlotte Warren-Gash
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
47
|
Ungvari Z, Toth P, Tarantini S, Prodan CI, Sorond F, Merkely B, Csiszar A. Hypertension-induced cognitive impairment: from pathophysiology to public health. Nat Rev Nephrol 2021; 17:639-654. [PMID: 34127835 PMCID: PMC8202227 DOI: 10.1038/s41581-021-00430-6] [Citation(s) in RCA: 275] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
Hypertension affects two-thirds of people aged >60 years and significantly increases the risk of both vascular cognitive impairment and Alzheimer's disease. Hypertension compromises the structural and functional integrity of the cerebral microcirculation, promoting microvascular rarefaction, cerebromicrovascular endothelial dysfunction and neurovascular uncoupling, which impair cerebral blood supply. In addition, hypertension disrupts the blood-brain barrier, promoting neuroinflammation and exacerbation of amyloid pathologies. Ageing is characterized by multifaceted homeostatic dysfunction and impaired cellular stress resilience, which exacerbate the deleterious cerebromicrovascular effects of hypertension. Neuroradiological markers of hypertension-induced cerebral small vessel disease include white matter hyperintensities, lacunar infarcts and microhaemorrhages, all of which are associated with cognitive decline. Use of pharmaceutical and lifestyle interventions that reduce blood pressure, in combination with treatments that promote microvascular health, have the potential to prevent or delay the pathogenesis of vascular cognitive impairment and Alzheimer's disease in patients with hypertension.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Farzaneh Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
48
|
Chifu I, Detomas M, Dischinger U, Kimpel O, Megerle F, Hahner S, Fassnacht M, Altieri B. Management of Patients With Glucocorticoid-Related Diseases and COVID-19. Front Endocrinol (Lausanne) 2021; 12:705214. [PMID: 34594302 PMCID: PMC8476969 DOI: 10.3389/fendo.2021.705214] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is a global health crisis affecting millions of people worldwide. SARS-CoV-2 enters the host cells by binding to angiotensin-converting enzyme 2 (ACE2) after being cleaved by the transmembrane protease serine 2 (TMPRSS2). In addition to the lung, gastrointestinal tract and kidney, ACE2 is also extensively expressed in endocrine tissues, including the pituitary and adrenal glands. Although glucocorticoids could play a central role as immunosuppressants during the cytokine storm, they can have both stimulating and inhibitory effects on immune response, depending on the timing of their administration and their circulating levels. Patients with adrenal insufficiency (AI) or Cushing's syndrome (CS) are therefore vulnerable groups in relation to COVID-19. Additionally, patients with adrenocortical carcinoma (ACC) could also be more vulnerable to COVID-19 due to the immunosuppressive state caused by the cancer itself, by secreted glucocorticoids, and by anticancer treatments. This review comprehensively summarizes the current literature on susceptibility to and outcome of COVID-19 in AI, CS and ACC patients and emphasizes potential pathophysiological mechanisms of susceptibility to COVID-19 as well as the management of these patients in case of SARS-CoV-2. Finally, by performing an in silico analysis, we describe the mRNA expression of ACE2, TMPRSS2 and the genes encoding their co-receptors CTSB, CTSL and FURIN in normal adrenal and adrenocortical tumors (both adenomas and carcinomas).
Collapse
Affiliation(s)
- Irina Chifu
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Mario Detomas
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Ulrich Dischinger
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Otilia Kimpel
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Felix Megerle
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Stefanie Hahner
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
- Central Laboratory, University Hospital Würzburg, Würzburg, Germany
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| |
Collapse
|
49
|
Memiah P, Nkinda L, Majigo M, Opanga Y, Humwa F, Inzaule S, Abubakar M, Oduor P, Zuheri A, Lema S, Kamau A, Baribwira C, Biadgilign S. Hypertension and Associated Inflammatory Markers Among HIV-Infected Patients in Tanzania. J Interferon Cytokine Res 2021; 41:291-301. [PMID: 34410876 DOI: 10.1089/jir.2021.0059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
There remains a dearth of data regarding the association between chronic inflammation and hypertension (HTN) in sub-Saharan Africa, a region that accounts for >70% of the global burden of HIV infection. Therefore, we assessed the levels of biomarkers among HIV+ individuals and its associations with HTN in Tanzania. A cross-sectional study was conducted at one of the largest clinics in Tanzania and data from 261 HIV+ patients were analyzed. Standardized tools were used to collect data. Blood pressure was measured using Omron® M2 blood pressure monitor. Enzyme-linked immunosorbent assay was used to test for inflammatory markers [C-reactive protein (CRP), interleukin (IL)-6, IL-18, soluble tumor necrosis factor receptor type I (sTNFRI), sTNFRII]. Bivariate and multivariable analysis was conducted to examine association between the biomarkers and HTN. We further conducted age-sex-alcohol-adjusted models to control for any confounders. The prevalence of HTN was 43% with a high prevalence reported in female (70%) participants and those older than 55 years of age (77%). Being women, older than 55 years of age, married, and being overweight was associated with HTN. The highest correlations were observed between TNR2 and CRP (ɤ = 0.13, P = 0.044), and TNR2 and IL-18 (ɤ = 0.13, P = 0.034). Participants who had elevated CRP levels were 2 times more likely to experience HTN in the age-adjusted model [odds ratio (OR) = 3.5, 95% confidence interval (CI) = 1.1-11.3], age-sex-adjusted model (OR = 3.3, 95% CI = 1.0-10.9), and the full model (OR = 2.9, 95% CI = 0.8-10.0). Our study shows that high CRP levels are significantly associated with the higher prevalence of HTN notwithstanding all other markers, which showed a positive association with HTN despite not being significant. These findings point to the importance of creating awareness, education, and screening for HTN among HIV patients in high epidemic countries. More rigorous studies are needed to know the exact pathway mechanisms of inflammation in HIV patients.
Collapse
Affiliation(s)
- Peter Memiah
- Division of Epidemiology and Prevention, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lillian Nkinda
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, United Republic of Tanzania
| | - Mtebe Majigo
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, United Republic of Tanzania
| | - Yvonne Opanga
- Department of Monitoring Evaluation and Research, Amref Health Africa, Nairobi, Kenya
| | - Felix Humwa
- Global Program for Research Teaching-University of California San Francisco, Nairobi, Kenya
| | - Seth Inzaule
- Department of Global Health, Amsterdam Institute for Global Health and Development, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Maghimbi Abubakar
- Center for International Health, Education, and Biosecurity (CIHEB), Institute of Human Virology, University of Maryland School of Medicine in Tanzania, Dar es Salaam, Tanzania
| | - Patience Oduor
- Center for International Health, Education, and Biosecurity (CIHEB), Institute of Human Virology, University of Maryland School of Medicine in Tanzania, Dar es Salaam, Tanzania
| | - Aisha Zuheri
- Infectious Disease Centre, Tanzania, Dar es Salaam, Tanzania
| | - Steven Lema
- Department of Global Health, Amsterdam Institute for Global Health and Development, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne Kamau
- Institute for Development Studies, University of Nairobi, Nairobi, Kenya
| | - Cyprien Baribwira
- PUniversity of Maryland School of Medicine in Rwanda, Kigali, Rwanda
| | | |
Collapse
|
50
|
Das UN. Molecular biochemical aspects of salt (sodium chloride) in inflammation and immune response with reference to hypertension and type 2 diabetes mellitus. Lipids Health Dis 2021; 20:83. [PMID: 34334139 PMCID: PMC8327432 DOI: 10.1186/s12944-021-01507-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 07/14/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity, insulin resistance, type 2 diabetes mellitus (T2DM) and hypertension (HTN) are common that are associated with low-grade systemic inflammation. Diet, genetic factors, inflammation, and immunocytes and their cytokines play a role in their pathobiology. But the exact role of sodium, potassium, magnesium and other minerals, trace elements and vitamins in the pathogenesis of HTN and T2DM is not known. Recent studies showed that sodium and potassium can modulate oxidative stress, inflammation, alter the autonomic nervous system and induce dysfunction of the innate and adaptive immune responses in addition to their action on renin-angiotensin-aldosterone system. These actions of sodium, potassium and magnesium and other minerals, trace elements and vitamins are likely to be secondary to their action on pro-inflammatory cytokines IL-6, TNF-α and IL-17 and metabolism of essential fatty acids that may account for their involvement in the pathobiology of insulin resistance, T2DM, HTN and autoimmune diseases.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St, Battle Ground, WA, 98604, USA.
| |
Collapse
|