RESUMEN
Our brain is not an immune-privileged island isolated from peripheries, but how non-neuronal brain cells interact with the peripheral system is not well understood. Wei et al. report that microglia in the hypothalamic paraventricular nucleus (PVN) with unique vasculature can detect ATP derived from hemodynamic disturbance. These microglia in the PVN regulate the response to hypertension via ATP-P2Y12-C/EBPß signaling.
Asunto(s)
Presión Sanguínea , Encéfalo , Microglía , Núcleo Hipotalámico Paraventricular , Microglía/inmunología , Microglía/fisiología , Microglía/metabolismo , Animales , Humanos , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/inmunología , Núcleo Hipotalámico Paraventricular/fisiología , Presión Sanguínea/fisiología , Encéfalo/inmunología , Adenosina Trifosfato/metabolismo , Transducción de Señal , Hipertensión/inmunología , Hipertensión/fisiopatología , Proteína beta Potenciadora de Unión a CCAAT/metabolismoRESUMEN
Hypertension affects one-third of the world's population, leading to cardiac dysfunction that is modulated by resident and recruited immune cells. Cardiomyocyte growth and increased cardiac mass are essential to withstand hypertensive stress; however, whether immune cells are involved in this compensatory cardioprotective process is unclear. In normotensive animals, single-cell transcriptomics of fate-mapped self-renewing cardiac resident macrophages (RMs) revealed transcriptionally diverse cell states with a core repertoire of reparative gene programs, including high expression of insulin-like growth factor-1 (Igf1). Hypertension drove selective in situ proliferation and transcriptional activation of some cardiac RM states, directly correlating with increased cardiomyocyte growth. During hypertension, inducible ablation of RMs or selective deletion of RM-derived Igf1 prevented adaptive cardiomyocyte growth, and cardiac mass failed to increase, which led to cardiac dysfunction. Single-cell transcriptomics identified a conserved IGF1-expressing macrophage subpopulation in human cardiomyopathy. Here we defined the absolute requirement of RM-produced IGF-1 in cardiac adaptation to hypertension.
Asunto(s)
Adaptación Fisiológica/fisiología , Hipertensión/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Macrófagos/metabolismo , Remodelación Ventricular/fisiología , Animales , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Hipertensión/complicaciones , Hipertensión/inmunología , Lactante , Masculino , Ratones , Persona de Mediana Edad , Miocardio/inmunología , Miocardio/metabolismo , Miocardio/patologíaRESUMEN
The SARS-CoV-2 pandemic has resulted in millions of infections, yet the role of host immune responses in early COVID-19 pathogenesis remains unclear. By investigating 17 acute and 24 convalescent patients, we found that acute SARS-CoV-2 infection resulted in broad immune cell reduction including T, natural killer, monocyte, and dendritic cells (DCs). DCs were significantly reduced with functional impairment, and ratios of conventional DCs to plasmacytoid DCs were increased among acute severe patients. Besides lymphocytopenia, although neutralizing antibodies were rapidly and abundantly generated in patients, there were delayed receptor binding domain (RBD)- and nucleocapsid protein (NP)-specific T cell responses during the first 3 weeks after symptoms onset. Moreover, acute RBD- and NP-specific T cell responses included relatively more CD4 T cells than CD8 T cells. Our findings provided evidence that impaired DCs, together with timely inverted strong antibody but weak CD8 T cell responses, could contribute to acute COVID-19 pathogenesis and have implications for vaccine development.
Asunto(s)
Betacoronavirus/patogenicidad , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Coronavirus/inmunología , Células Dendríticas/inmunología , Diabetes Mellitus/inmunología , Hipertensión/inmunología , Neumonía Viral/inmunología , Adulto , Anciano , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Betacoronavirus/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , COVID-19 , Convalecencia , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/virología , Células Dendríticas/patología , Células Dendríticas/virología , Complicaciones de la Diabetes , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/virología , Progresión de la Enfermedad , Femenino , Humanos , Hipertensión/complicaciones , Hipertensión/diagnóstico , Hipertensión/virología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Células Asesinas Naturales/virología , Activación de Linfocitos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/patología , Monocitos/virología , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/diagnóstico , Neumonía Viral/virología , SARS-CoV-2 , Índice de Severidad de la EnfermedadRESUMEN
Obesity is increasingly prevalent and is associated with substantial cardiovascular risk. Adipose tissue distribution and morphology play a key role in determining the degree of adverse effects, and a key factor in the disease process appears to be the inflammatory cell population in adipose tissue. Healthy adipose tissue secretes a number of vasoactive adipokines and anti-inflammatory cytokines, and changes to this secretory profile will contribute to pathogenesis in obesity. In this review, we discuss the links between adipokine dysregulation and the development of hypertension and diabetes and explore the potential for manipulating adipose tissue morphology and its immune cell population to improve cardiovascular health in obesity.
Asunto(s)
Tejido Adiposo/fisiopatología , Presión Sanguínea , Diabetes Mellitus/fisiopatología , Hipertensión/fisiopatología , Obesidad/fisiopatología , Adipoquinas/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Adiposidad , Animales , Diabetes Mellitus/epidemiología , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Humanos , Hipertensión/epidemiología , Hipertensión/inmunología , Hipertensión/metabolismo , Mediadores de Inflamación/metabolismo , Obesidad/epidemiología , Obesidad/inmunología , Obesidad/metabolismo , Fenotipo , Medición de Riesgo , Factores de Riesgo , Transducción de Señal , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatologíaRESUMEN
BACKGROUND: Hypertension is characterized by CD8+ (cluster differentiation 8) T cell activation and infiltration into peripheral tissues. CD8+ T cell activation requires proteasomal processing of antigenic proteins. It has become clear that isoLG (isolevuglandin)-adduced peptides are antigenic in hypertension; however, IsoLGs inhibit the constitutive proteasome. We hypothesized that immunoproteasomal processing of isoLG-adducts is essential for CD8+ T cell activation and inflammation in hypertension. METHODS: IsoLG adduct processing was studied in murine dendritic cells (DCs), endothelial cells (ECs), and B8 fibroblasts. The role of the proteasome and the immunoproteasome in Ang II (angiotensin II)-induced hypertension was studied in C57BL/6 mice treated with bortezomib or the immunoproteasome inhibitor PR-957 and by studying mice lacking 3 critical immunoproteasome subunits (triple knockout mouse). We also examined hypertension in mice lacking the critical immunoproteasome subunit LMP7 (large multifunctional peptidase 7) specifically in either DCs or ECs. RESULTS: We found that oxidant stress increases the presence of isoLG adducts within MHC-I (class I major histocompatibility complex), and immunoproteasome overexpression augments this. Pharmacological or genetic inhibition of the immunoproteasome attenuated hypertension and tissue inflammation. Conditional deletion of LMP7 in either DCs or ECs attenuated hypertension and vascular inflammation. Finally, we defined the role of the innate immune receptors STING (stimulator of interferon genes) and TLR7/8 (toll-like receptor 7/8) as drivers of LMP7 expression in ECs. CONCLUSIONS: These studies define a previously unknown role of the immunoproteasome in DCs and ECs in CD8+ T cell activation. The immunoproteasome in DCs and ECs is critical for isoLG-adduct presentation to CD8+ T cells, and in the endothelium, this guides homing and infiltration of T cells to specific tissues.
Asunto(s)
Bortezomib , Linfocitos T CD8-positivos , Células Dendríticas , Hipertensión , Complejo de la Endopetidasa Proteasomal , Animales , Masculino , Ratones , Angiotensina II , Bortezomib/farmacología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/inmunología , Fibroblastos/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Hipertensión/metabolismo , Hipertensión/inmunología , Activación de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Oligopéptidos , Estrés Oxidativo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacologíaRESUMEN
The brain is essential for processing and integrating sensory signals coming from peripheral tissues. Conversely, the autonomic nervous system regulated by brain centres modulates the immune responses involved in the genesis and progression of cardiovascular diseases. Understanding the pathophysiological bases of this relationship established between the brain and immune system is relevant for advancing therapies. An additional mechanism involved in the regulation of cardiovascular function is provided by the brain-mediated control of the renin-angiotensin system. In both cases, the communication is typically bidirectional and established by afferent and sensory signals collected at the level of peripheral tissues, efferent circuits, as well as of hormones. Understanding how the brain mediates the bidirectional communication and how the immune system participates in this process is object of intense investigation. This review examines key findings that support a role for these interactions in the pathogenesis of major vascular diseases that are characterized by a consistent alteration of the immune response, such as hypertension and atherosclerosis. In addition, we provide a critical appraisal of the translational implications that these discoveries have in the clinical setting where an effective management of neuroimmune and/or neuroinflammatory state might be beneficial.
Asunto(s)
Aterosclerosis , Hipertensión , Neuroinmunomodulación , Humanos , Hipertensión/inmunología , Hipertensión/fisiopatología , Animales , Aterosclerosis/inmunología , Neuroinmunomodulación/fisiología , Encéfalo/inmunología , Sistema Renina-Angiotensina/fisiología , Sistema Nervioso Autónomo/fisiopatología , Sistema Nervioso Autónomo/inmunologíaRESUMEN
High blood pressure is present in more than one billion adults worldwide and is the most important modifiable risk factor of death resulting from cardiovascular disease. While many factors contribute to the pathogenesis of hypertension, a role of the immune system has been firmly established by a large number of investigations from many laboratories around the world. Immunosuppressive drugs and inhibition of individual cytokines prevent or ameliorate experimental hypertension, and studies in genetically-modified mouse strains have demonstrated that lymphocytes are necessary participants in the development of hypertension and in hypertensive organ injury. Furthermore, immune reactivity may be the driving force of hypertension in autoimmune diseases. Infiltration of immune cells, oxidative stress, and stimulation of the intrarenal angiotensin system are induced by activation of the innate and adaptive immunity. High blood pressure results from the combined effects of inflammation-induced impairment in the pressure natriuresis relationship, dysfunctional vascular relaxation, and overactivity of the sympathetic nervous system. Imbalances between proinflammatory effector responses and anti-inflammatory responses of regulatory T cells to a large extent determine the severity of inflammation. Experimental and human studies have uncovered autoantigens (isoketal-modified proteins and heat shock protein 70) of potential clinical relevance. Further investigations on the immune reactivity in hypertension may result in the identification of new strategies for the treatment of the disease.
Asunto(s)
Presión Sanguínea/inmunología , Citocinas/inmunología , Hipertensión/inmunología , Inmunidad Adaptativa , Animales , Autoantígenos/inmunología , Autoinmunidad , Bacterias/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Interacciones Huésped-Patógeno , Humanos , Hipertensión/metabolismo , Hipertensión/microbiología , Hipertensión/fisiopatología , Inmunidad Innata , Factores de Riesgo , Transducción de SeñalRESUMEN
We used an animal model of salt-sensitive hypertension (SSH) in which ovariectomized (oVx) rats developed hypertension with high salt (HS) intake. Hypertension is accompanied by changes in the percentage of CD4+ T lymphocytes, immune CD45+ cell infiltration into renal tissue, and changes in Na+, K+- ATPase (NKA) expression in both renal tissue and peripheral blood mononuclear cells (PBMCs). To determine whether the observed changes resulted from HS intake, high blood pressure, or both, hydralazine (HDZ) was used to lower blood pressure. The oVx HS rats received two HDZ schedules either to prevent or to treat hypertension. NKA was overexpressed in the kidneys of all oVx groups and in PBMCs of oVx HS rats. This pattern was not altered with HDZ treatment. Changes in CD4+ T lymphocytes and renal infiltration of CD45+ cells were not reversed either. High salt, but not high blood pressure, induces immune cell activation and renal infiltration. Overexpressed NKA is the primary event, and HS is the perturbation to the system in this model of SSH, which resembles the postmenopausal state.
Asunto(s)
Hipertensión , Riñón , Ovariectomía , Ratas Wistar , Animales , Femenino , Ratas , Riñón/patología , Riñón/metabolismo , Riñón/inmunología , Hipertensión/inmunología , Hipertensión/patología , Hipertensión/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Presión Sanguínea/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Hidralazina/farmacologíaRESUMEN
CD40L-CD40-TRAF signaling plays a role in atherosclerosis progression and affects the pathogenesis of coronary heart disease (CHD). We tested the hypothesis that CD40L-CD40-TRAF signaling is a potential therapeutic target in hyperlipidemia, diabetes, and hypertension. In mouse models of hyperlipidemia plus diabetes (db/db mice) or hypertension (1 mg/kg/d angiotensin-II for 7 days), TRAF6 inhibitor treatment (2.5 mg/kg/d for 7 or 14 days) normalized markers of oxidative stress and inflammation. As diabetes and hypertension are important comorbidities aggravating CHD, we explored whether the CD40L-CD40-TRAF signaling cascade and their associated inflammatory pathways are expressed in CHD patients suffering from comorbidities. Therefore, we analyzed vascular bypass material (aorta or internal mammary artery) and plasma from patients with CHD with diabetes and/or hypertension. Our Olink targeted plasma proteomic analysis using the IMMUNO-ONCOLOGY panel revealed a pattern of step-wise increase for 13/92 markers of low-grade inflammation with significant changes. CD40L or CD40 significantly correlated with 38 or 56 other inflammatory targets. In addition, specific gene clusters that correlate with the comorbidities were identified in isolated aortic mRNA of CHD patients through RNA-sequencing. These signaling clusters comprised CD40L-CD40-TRAF, immune system, hemostasis, muscle contraction, metabolism of lipids, developmental biology, and apoptosis. Finally, immunological analysis revealed key markers correlated with comorbidities in CHD patients, such as CD40L, NOX2, CD68, and 3-nitrotyrosine. These data indicate that comorbidities increase inflammatory pathways in CHD, and targeting these pathways will be beneficial in reducing cardiovascular events in CHD patients with comorbidities.
Asunto(s)
Antígenos CD40 , Ligando de CD40 , Hipertensión , Transducción de Señal , Humanos , Animales , Ligando de CD40/metabolismo , Hipertensión/inmunología , Hipertensión/metabolismo , Antígenos CD40/metabolismo , Masculino , Inflamación/metabolismo , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Femenino , Persona de Mediana Edad , Factor 6 Asociado a Receptor de TNF/metabolismo , Anciano , Enfermedad Coronaria/inmunología , Enfermedad Coronaria/metabolismoRESUMEN
Salt-sensitive hypertension (SSHTN) is associated with M1 macrophage polarization and inflammatory responses, leading to inflammation-associated lymphangiogenesis and functional impairment across multiple organs, including kidneys and gonads. However, it remains unclear whether promoting M2 macrophage polarization can alleviate the hypertension, inflammation, and end organ damage in mice with salt sensitive hypertension (SSHTN). Male and female mice were made hypertensive by administering nitro-L-arginine methyl ester hydrochloride (L-NAME; 0.5 mg/ml) for 2 weeks in the drinking water, followed by a 2-week interval without any treatments, and a subsequent high salt diet for 3 weeks (SSHTN). AVE0991 (AVE) was intraperitoneally administered concurrently with the high salt diet. Control mice were provided standard diet and tap water. AVE treatment significantly attenuated BP and inflammation in mice with SSHTN. Notably, AVE promoted M2 macrophage polarization, decreased pro-inflammatory immune cell populations, and improved function in renal and gonadal tissues of mice with SSHTN. Additionally, AVE decreased lymphangiogenesis in the kidneys and testes of male SSHTN mice and the ovaries of female SSHTN mice. These findings highlight the effectiveness of AVE in mitigating SSHTN-induced elevated BP, inflammation, and end organ damage by promoting M2 macrophage polarization and suppressing pro-inflammatory immune responses. Targeting macrophage polarization emerges as a promising therapeutic approach for alleviating inflammation and organ damage in SSHTN. Further studies are warranted to elucidate the precise mechanisms underlying AVE-mediated effects and to assess its clinical potential in managing SSHTN.
Asunto(s)
Hipertensión , Inflamación , Riñón , Macrófagos , Cloruro de Sodio Dietético , Animales , Masculino , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Femenino , Hipertensión/inmunología , Hipertensión/tratamiento farmacológico , Hipertensión/fisiopatología , Riñón/efectos de los fármacos , Riñón/patología , Riñón/inmunología , Linfangiogénesis/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones , Presión Sanguínea/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/patología , Modelos Animales de EnfermedadRESUMEN
We reported that salt-sensitive hypertension (SSHTN) is associated with increased pro-inflammatory immune cells, inflammation, and inflammation-associated lymphangiogenesis in the kidneys and gonads of male and female mice. However, it is unknown whether these adverse end organ effects result from increased blood pressure (BP), elevated levels of salt, or both. We hypothesized that pharmaceutically lowering BP would not fully alleviate the renal and gonadal immune cell accumulation, inflammation, and lymphangiogenesis associated with SSHTN. SSHTN was induced in male and female C57BL6/J mice by administering nitro-L-arginine methyl ester hydrochloride (L-NAME; 0.5 mg/ml) in their drinking water for 2 weeks, followed by a 2-week washout period. Subsequently, the mice received a 3-week 4% high salt diet (SSHTN). The treatment group underwent the same SSHTN induction protocol but received hydralazine (HYD; 250 mg/L) in their drinking water during the diet phase (SSHTN+HYD). Control mice received tap water and a standard diet for 7 weeks. In addition to decreasing systolic BP, HYD treatment generally decreased pro-inflammatory immune cells and inflammation in the kidneys and gonads of SSHTN mice. Furthermore, the decrease in BP partially alleviated elevated renal and gonadal lymphatics and improved renal and gonadal function in mice with SSHTN. These data demonstrate that high systemic pressure and salt differentially act on end organ immune cells, contributing to the broader understanding of how BP and salt intake collectively shape immune responses and highlight implications for targeted therapeutic interventions.
Asunto(s)
Presión Sanguínea , Hipertensión , Inflamación , Riñón , Ratones Endogámicos C57BL , Cloruro de Sodio Dietético , Animales , Hipertensión/inmunología , Hipertensión/fisiopatología , Hipertensión/tratamiento farmacológico , Hipertensión/inducido químicamente , Masculino , Femenino , Presión Sanguínea/efectos de los fármacos , Cloruro de Sodio Dietético/efectos adversos , Riñón/inmunología , Riñón/efectos de los fármacos , Inflamación/inmunología , Linfangiogénesis/efectos de los fármacos , Antihipertensivos/farmacología , Ratones , Hidralazina/farmacología , NG-Nitroarginina Metil Éster/farmacología , Modelos Animales de Enfermedad , Gónadas/efectos de los fármacosRESUMEN
OBJECTIVE: The prevalence of hypertension, a major cardiovascular risk factor, is increased in patients with rheumatoid arthritis (RA) and may be driven by immune activation. The purpose of this study was to determine if ambulatory 24-hour blood pressure (BP) is elevated in RA vs control participants and whether it is associated with immune activation. METHODS: We conducted a cross-sectional study of 46 patients with RA and 23 control participants. Participants wore an ambulatory BP monitor that obtained diurnal BP every 15-30 minutes and nocturnal BP every 30 minutes. Inflammatory mediators in plasma were measured using an inflammation proteomics panel. Differences in BP measurements were assessed by Mann-Whitney U test, and association with inflammatory mediators was assessed by Spearman correlation. RESULTS: Patients with RA and control participants had similar office BP, but median ambulatory systolic BP (SBP) measurements (24-hour [RA 121 mmHg vs control 116 mmHg; P = 0.01], diurnal [RA 128 mmHg vs control 120 mmHg; P = 0.003], and nocturnal [RA 112 mmHg vs control 103 mmHg; P = 0.002]) were higher in patients with RA. Patients with RA also had higher nocturnal diastolic BP (DBP; RA 63 mmHg vs control 57 mmHg; P = 0.02), but other DBP measurements were similar. Nocturnal BP dipping was less in patients with RA (12%) compared to control participants (16%; P = 0.02). In patients with RA, higher 24-hour and nocturnal SBPs and less nocturnal dipping were strongly correlated with a wide range of inflammatory mediators. CONCLUSION: Despite similar office measurements, 24-hour and nocturnal SBP measurements were higher in patients with RA than in control participants and were strongly associated with inflammation.
Asunto(s)
Artritis Reumatoide , Monitoreo Ambulatorio de la Presión Arterial , Presión Sanguínea , Hipertensión , Humanos , Artritis Reumatoide/inmunología , Artritis Reumatoide/fisiopatología , Artritis Reumatoide/sangre , Femenino , Masculino , Persona de Mediana Edad , Estudios Transversales , Presión Sanguínea/fisiología , Hipertensión/fisiopatología , Hipertensión/inmunología , Anciano , Adulto , Ritmo Circadiano/fisiologíaRESUMEN
PURPOSE: This review aimed to investigate the prevalence of hypertension and cardiovascular (CV) complications in various inflammatory and autoimmune diseases (IAD). RECENT FINDINGS: Despite recent improvements in the management of IAD, patients with IAD still have an increased CV mortality and CV complications, mostly related to CV risk factors such as hypertension and inflammation. We systematically searched MEDLINE and EMBASE libraries for controlled studies involving hypertension and CV complications in systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), psoriasis including psoriatic arthritis (PsA), Sjogren's syndrome (SS), or antineutrophil cytoplasmic antibody-associated vasculitis (AAV) between January 2000 and March 2022. We extracted data on the prevalence of hypertension and CV complications. Then, random-effects meta-analyses and exploratory multivariate meta-regression were performed to explore factors related to the prevalence of hypertension. Of 2726 studies screened, 122 were selected for the meta-analysis. The prevalence of hypertension was higher among patients with IAD than controls, with an overall unadjusted odds ratio (OR) [95% confidence interval] of 1.67 [1.58-1.76] and an adjusted OR of 1.36 [1.24-1.50]. All diseases were found to be associated with increased risk of hypertension: SLE, adjusted OR 3.40 [1.93-6.00]; psoriasis, OR 1.32 [1.16-1.51]; PsA, OR 1.49 [1.15-1.94]; RA, OR 1.28 [1.04-1.58]; SS, OR 2.02 [1.19-3.44]. Age and female sex were significantly associated with hypertension in patients with IAD. The risk of CV complications was increased: ischemic heart disease, adjusted OR 1.38 [1.21-1.57]; cerebrovascular disease, OR 1.37 [1.03-1.81]; heart failure, OR 1.28 [1.05-1.55]; atherosclerotic plaques presence, OR 2.46 [1.84-3.29]. The prevalence of hypertension and CV complications is higher among patients with IAD. Screening and management of hypertension appears to be of paramount importance in these patients.
Asunto(s)
Enfermedades Autoinmunes , Enfermedades Cardiovasculares , Hipertensión , Humanos , Enfermedades Autoinmunes/complicaciones , Enfermedades Autoinmunes/epidemiología , Enfermedades Autoinmunes/inmunología , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/inmunología , Hipertensión/epidemiología , Hipertensión/inmunología , Inflamación , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/epidemiología , Prevalencia , Factores de RiesgoRESUMEN
BACKGROUND: The pathogenesis of hypertension (HTN) in people living with HIV/AIDS (PLHIV) is complex and remains not fully understood. Chronic immune activation (IA) is postulated to be one of the culprits. This notion is derived from studies in HIV-uninfected populations and/or animals while data on HTN and how it relates to IA in PLHIV remains scarce. We determined the relationship between HTN and IA among antiretroviral therapy (ART) naïve PLHIV. METHODS: We analysed baseline data of 365 out of 430 clinical trial participants whose main aim was to investigate the effect of low-dose aspirin on HIV disease progression in PLHIV starting ART. Soluble CD14 (sCD14), T cells co-expressing CD38 and HLA-DR, and PD-1 were the IA and exhaustion markers, respectively studied and were analysed by flow cytometry. Mann-Whitney U-test was used for comparison of the markers by HTN status. A robust Poisson regression model was used to determine the predictors for HTN. RESULTS: A quarter of the 365 were hypertensive (25.3%, 95% CI 20.9-29.8%), and, had higher median (IQR) body mass index (kg/m2) (23.4 (19.6, 28.0) versus 21.9 (19.3, 25.1)) and lower median (IQR) estimated glomerular filtration rate (mL/min/1.73m2) (101.2 (79.4, 126.9) versus 113.6 (92.7, 138.8)) than normotensive participants (p < 0.05). Participants with HTN had higher median frequencies of all markers of IA and exhaustion but lower sCD14 (p > 0.05). None of these markers significantly predicted the occurrence of HTN. CONCLUSION: Studied markers of IA and exhaustion were higher in PLHIV with HTN than those without but were unpredictive of HTN. Larger multicentre studies with a wider range of markers are needed to confirm the role of IA in HIV-associated HTN.
Asunto(s)
Infecciones por VIH , Hipertensión , Humanos , Masculino , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Infecciones por VIH/complicaciones , Femenino , Adulto , Hipertensión/tratamiento farmacológico , Hipertensión/inmunología , Persona de Mediana Edad , Receptores de Lipopolisacáridos/sangre , Biomarcadores/sangreRESUMEN
Immune response dysregulation plays a key role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogenesis. In this study, we evaluated immune and endothelial blood cell profiles of patients with coronavirus disease 2019 (COVID-19) to determine critical differences between those with mild, moderate, or severe COVID-19 using spectral flow cytometry. We examined a suite of immune phenotypes, including monocytes, T cells, NK cells, B cells, endothelial cells, and neutrophils, alongside surface and intracellular markers of activation. Our results showed progressive lymphopenia and depletion of T cell subsets (CD3+, CD4+, and CD8+) in patients with severe disease and a significant increase in the CD56+CD14+Ki67+IFN-γ+ monocyte population in patients with moderate and severe COVID-19 that has not been previously described. Enhanced circulating endothelial cells (CD45-CD31+CD34+CD146+), circulating endothelial progenitors (CD45-CD31+CD34+/-CD146-), and neutrophils (CD11b+CD66b+) were coevaluated for COVID-19 severity. Spearman correlation analysis demonstrated the synergism among age, obesity, and hypertension with upregulated CD56+ monocytes, endothelial cells, and decreased T cells that lead to severe outcomes of SARS-CoV-2 infection. Circulating monocytes and endothelial cells may represent important cellular markers for monitoring postacute sequelae and impacts of SARS-CoV-2 infection during convalescence and for their role in immune host defense in high-risk adults after vaccination.
Asunto(s)
COVID-19/inmunología , Células Endoteliales/inmunología , Monocitos/inmunología , SARS-CoV-2 , Adolescente , Adulto , Factores de Edad , Anciano , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Biomarcadores , Antígeno CD56/análisis , COVID-19/sangre , COVID-19/epidemiología , Niño , Comorbilidad , Células Endoteliales/química , Femenino , Citometría de Flujo , Humanos , Hipertensión/epidemiología , Hipertensión/inmunología , Inmunofenotipificación , Activación de Linfocitos , Subgrupos Linfocitarios/inmunología , Linfopenia/etiología , Linfopenia/inmunología , Masculino , Persona de Mediana Edad , Monocitos/química , Neutrófilos/inmunología , Obesidad/epidemiología , Obesidad/inmunología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad , Glicoproteína de la Espiga del Coronavirus/inmunología , Adulto JovenRESUMEN
T cell depletion can prevent hypertension in experimental animals. What is the nature of T cell activation in hypertension? In this issue of Immunity, Carnevale et al. (2014) implicate PlGF signaling in a reservoir of splenic T cells.
Asunto(s)
Presión Sanguínea/inmunología , Hipertensión/inmunología , Proteínas Gestacionales/inmunología , Bazo/inmunología , Animales , Factor de Crecimiento PlacentarioRESUMEN
Hypertension is a health problem affecting over 1 billion people worldwide. How the immune system gets activated under hypertensive stimuli to contribute to blood pressure elevation is a fascinating enigma. Here we showed a splenic role for placental growth factor (PlGF), which accounts for the onset of hypertension, through immune system modulation. PlGF repressed the expression of the protein Timp3 (tissue inhibitor of metalloproteinases 3), through the transcriptional Sirt1-p53 axis. Timp3 repression allowed costimulation of T cells and their deployment toward classical organs involved in hypertension. We showed that the spleen is an essential organ for the development of hypertension through a noradrenergic drive mediated by the celiac ganglion efferent. Overall, we demonstrate that PlGF mediates the neuroimmune interaction in the spleen, organizing a unique and nonredundant response that allows the onset of hypertension.
Asunto(s)
Presión Sanguínea/inmunología , Hipertensión/inmunología , Proteínas Gestacionales/inmunología , Bazo/inmunología , Angiotensina II/inmunología , Animales , Presión Sanguínea/genética , Ganglios Simpáticos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroinmunomodulación , Factor de Crecimiento Placentario , Proteínas Gestacionales/genética , Interferencia de ARN , ARN Interferente Pequeño , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Linfocitos T/inmunología , Inhibidor Tisular de Metaloproteinasa-3/biosíntesis , Inhibidor Tisular de Metaloproteinasa-3/genética , Proteína p53 Supresora de Tumor/genéticaRESUMEN
Elevated cardiovascular risk including stroke, heart failure, and heart attack is present even after normalization of blood pressure in patients with hypertension. Underlying immune cell activation is a likely culprit. Although immune cells are important for protection against invading pathogens, their chronic overactivation may lead to tissue damage and high blood pressure. Triggers that may initiate immune activation include viral infections, autoimmunity, and lifestyle factors such as excess dietary salt. These conditions activate the immune system either directly or through their impact on the gut microbiome, which ultimately produces chronic inflammation and hypertension. T cells are central to the immune responses contributing to hypertension. They are activated in part by binding specific antigens that are presented in major histocompatibility complex molecules on professional antigen-presenting cells, and they generate repertoires of rearranged T-cell receptors. Activated T cells infiltrate tissues and produce cytokines including interleukin 17A, which promote renal and vascular dysfunction and end-organ damage leading to hypertension. In this comprehensive review, we highlight environmental, genetic, and microbial associated mechanisms contributing to both innate and adaptive immune cell activation leading to hypertension. Targeting the underlying chronic immune cell activation in hypertension has the potential to mitigate the excess cardiovascular risk associated with this common and deadly disease.
Asunto(s)
Hipertensión/inmunología , Inmunidad Celular/fisiología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Antihipertensivos/uso terapéutico , Linfocitos B/inmunología , Proteínas del Sistema Complemento/inmunología , Citocinas/inmunología , Células Dendríticas/inmunología , Resistencia a Medicamentos , Femenino , Microbioma Gastrointestinal/inmunología , Factores de Riesgo de Enfermedad Cardiaca , Interacciones Microbiota-Huesped , Humanos , Hipertensión/tratamiento farmacológico , Fenómenos del Sistema Inmunológico , Inmunidad Innata , Inflamasomas/inmunología , Inflamación/genética , Inflamación/inmunología , Macrófagos/inmunología , Masculino , Monocitos/inmunología , Factores Sexuales , Cloruro de Sodio Dietético/efectos adversos , Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Virosis/inmunologíaRESUMEN
BACKGROUND: Severe coronavirus disease 2019 (COVID-19) is characterized by impaired type I interferon activity and a state of hyperinflammation leading to acute respiratory distress syndrome. The complement system has recently emerged as a key player in triggering and maintaining the inflammatory state, but the role of this molecular cascade in severe COVID-19 is still poorly characterized. OBJECTIVE: We aimed at assessing the contribution of complement pathways at both the protein and transcriptomic levels. METHODS: To this end, we systematically assessed the RNA levels of 28 complement genes in the circulating whole blood of patients with COVID-19 and healthy controls, including genes of the alternative pathway, for which data remain scarce. RESULTS: We found differential expression of genes involved in the complement system, yet with various expression patterns: whereas patients displaying moderate disease had elevated expression of classical pathway genes, severe disease was associated with increased lectin and alternative pathway activation, which correlated with inflammation and coagulopathy markers. Additionally, properdin, a pivotal positive regulator of the alternative pathway, showed high RNA expression but was found at low protein concentrations in patients with a severe and critical disease, suggesting its deposition at the sites of complement activation. Notably, low properdin levels were significantly associated with the use of mechanical ventilation (area under the curve = 0.82; P = .002). CONCLUSION: This study sheds light on the role of the alternative pathway in severe COVID-19 and provides additional rationale for the testing of drugs inhibiting the alternative pathway of the complement system.
Asunto(s)
COVID-19/inmunología , Activación de Complemento/genética , Vía Alternativa del Complemento/genética , Proteínas del Sistema Complemento/genética , Coagulación Intravascular Diseminada/inmunología , SARS-CoV-2/patogenicidad , COVID-19/genética , COVID-19/terapia , COVID-19/virología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/terapia , Enfermedades Cardiovasculares/virología , Estudios de Casos y Controles , Comorbilidad , Proteínas del Sistema Complemento/inmunología , Diabetes Mellitus/genética , Diabetes Mellitus/inmunología , Diabetes Mellitus/terapia , Diabetes Mellitus/virología , Coagulación Intravascular Diseminada/genética , Coagulación Intravascular Diseminada/terapia , Coagulación Intravascular Diseminada/virología , Femenino , Regulación de la Expresión Génica , Humanos , Hipertensión/genética , Hipertensión/inmunología , Hipertensión/terapia , Hipertensión/virología , Lectinas/genética , Lectinas/inmunología , Masculino , Persona de Mediana Edad , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/virología , Properdina/genética , Properdina/inmunología , Respiración Artificial , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Índice de Severidad de la EnfermedadRESUMEN
Premenopausal females are protected from angiotensin II (ANG II)-induced hypertension following the adoptive transfer of T cells from normotensive donors. For the present study, we hypothesized that the transfer of hypertensive T cells (HT) or splenocytes (HS) from hypertensive donors would eliminate premenopausal protection from hypertension. Premenopausal recombination-activating gene-1 (Rag-1)-/- females received either normotensive (NT) or hypertensive cells 3 wk before ANG II infusion (14 days, 490 ng/kg/min). Contrary to our hypothesis, no increase in ANG II-induced blood pressure was observed in the NT/ANG or HT/ANG groups. Flow cytometry demonstrated that renal FoxP3+ T regulatory cells were significantly decreased, and immunohistochemistry showed an increase in renal F4/80+ macrophages in the HT/ANG group, suggesting a shift in the renal inflammatory environment despite no change in blood pressure. Renal mRNA expression of macrophage chemoattractant protein-1 (MCP-1), endothelin-1 (ET-1), and G protein-coupled estrogen receptor-1 (GPER-1) was significantly decreased in the HT/ANG group. The adoptive transfer of hypertensive splenocytes before ANG II infusion (HS/ANG) eliminated premenopausal protection from hypertension and significantly decreased splenic FoxP3+ T regulatory cells compared with females that received normotensive splenocytes (NS/ANG). Expression of macrophage inflammatory protein 1α/chemokine (C-C motif) ligand 3 (MCP-1/CCL3), a potent macrophage chemokine, was elevated in the HS/ANG group; however, no increase in renal macrophage infiltration occurred. Together, these data show that in premenopausal females, T cells from hypertensive donors are not sufficient to induce robust ANG II-mediated hypertension; in contrast, transfer of hypertensive splenocytes (consisting of T/B lymphocytes, dendritic cells, and macrophages) is sufficient. Further work is needed to understand how innate and adaptive immune cells and estrogen signaling coordinate to cause differential hypertensive outcomes in premenopausal females.NEW & NOTEWORTHY Our study is the first to explore the role of hypertensive T cells versus hypertensive splenocytes in premenopausal protection from ANG II-induced hypertension. We show that the hypertensive status of T cell donors does not impact blood pressure in the recipient female. However, splenocytes, when transferred from hypertensive donors, significantly increased premenopausal recipient blood pressure following ANG II infusion, highlighting the importance of further investigation into estrogen signaling and immune cell activation in females.