Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 131(9): 731-747, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36169218

RESUMEN

BACKGROUND: SH2B3 (SH2B adaptor protein 3) is an adaptor protein that negatively regulates cytokine signaling and cell proliferation. A common missense single nucleotide polymorphism in SH2B3 (rs3184504) results in substitution of tryptophan (Trp) for arginine (Arg) at amino acid 262 and is a top association signal for hypertension in human genome-wide association studies. Whether this variant is causal for hypertension, and if so, the mechanism by which it impacts pathogenesis is unknown. METHODS: We used CRISPR-Cas9 technology to create mice homozygous for the major (Arg/Arg) and minor (Trp/Trp) alleles of this SH2B3 polymorphism. Mice underwent angiotensin II (Ang II) infusion to evaluate differences in blood pressure (BP) elevation and end-organ damage including albuminuria and renal fibrosis. Cytokine production and Stat4 phosphorylation was also assessed in Arg/Arg and Trp/Trp T cells. RESULTS: Trp/Trp mice exhibit 10 mmHg higher systolic BP during chronic Ang II infusion compared to Arg/Arg controls. Renal injury and perivascular fibrosis are exacerbated in Trp/Trp mice compared to Arg/Arg controls following Ang II infusion. Renal and ex vivo stimulated splenic CD8+ T cells from Ang II-infused Trp/Trp mice produce significantly more interferon gamma (IFNg) compared to Arg/Arg controls. Interleukin-12 (IL-12)-induced IFNg production is greater in Trp/Trp compared to Arg/Arg CD8+ T cells. In addition, IL-12 enhances Stat4 phosphorylation to a greater degree in Trp/Trp compared to Arg/Arg CD8+ T cells, suggesting that Trp-encoding SH2B3 exhibits less negative regulation of IL-12 signaling to promote IFNg production. Finally, we demonstrated that a multi-SNP model genetically predicting increased SH2B3 expression in lymphocytes is inversely associated with hypertension and hypertensive chronic kidney disease in humans.. CONCLUSIONS: Taken together, these results suggest that the Trp encoding allele of rs3184504 is causal for BP elevation and renal dysfunction, in part through loss of SH2B3-mediated repression of T cell IL-12 signaling leading to enhanced IFNg production.


Asunto(s)
Hipertensión Renal , Hipertensión , Proteínas Adaptadoras Transductoras de Señales , Angiotensina II/metabolismo , Angiotensina II/toxicidad , Animales , Arginina/efectos adversos , Arginina/metabolismo , Linfocitos T CD8-positivos/metabolismo , Fibrosis , Estudio de Asociación del Genoma Completo , Humanos , Hipertensión/metabolismo , Hipertensión Renal/metabolismo , Interferón gamma/metabolismo , Interleucina-12/efectos adversos , Interleucina-12/metabolismo , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Triptófano
2.
Clin Sci (Lond) ; 137(16): 1225-1247, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37606086

RESUMEN

Heart failure with preserved ejection fraction (HFpEF) now accounts for the majority of new heart failure diagnoses and continues to increase in prevalence in the United States. Importantly, HFpEF is a highly morbid, heterogeneous syndrome lacking effective therapies. Inflammation has emerged as a potential contributor to the pathogenesis of HFpEF. Many of the risk factors for HFpEF are also associated with chronic inflammation, such as obesity, hypertension, aging, and renal dysfunction. A large amount of preclinical evidence suggests that immune cells and their associated cytokines play important roles in mediating fibrosis, oxidative stress, metabolic derangements, and endothelial dysfunction, all potentially important processes in HFpEF. How inflammation contributes to HFpEF pathogenesis, however, remains poorly understood. Recently, a variety of preclinical models have emerged which may yield much needed insights into the causal relationships between risk factors and the development of HFpEF, including the role of specific immune cell subsets or inflammatory pathways. Here, we review evidence in animal models and humans implicating inflammation as a mediator of HFpEF and identify gaps in knowledge requiring further study. As the understanding between inflammation and HFpEF evolves, it is hoped that a better understanding of the mechanisms underlying immune cell activation in HFpEF can open up new therapeutic avenues.


Asunto(s)
Insuficiencia Cardíaca , Animales , Humanos , Volumen Sistólico , Factores de Riesgo , Inflamación , Envejecimiento
3.
Circ Res ; 128(7): 908-933, 2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33793336

RESUMEN

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/inmunología
4.
Nutr Metab Cardiovasc Dis ; 33(7): 1398-1406, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37156670

RESUMEN

BACKGROUND AND AIMS: High sodium intake is associated with obesity and insulin resistance, and high extracellular sodium content may induce systemic inflammation, leading to cardiovascular disease. In this study, we aim to investigate whether high tissue sodium accumulation relates with obesity-related insulin resistance and whether the pro-inflammatory effects of excess tissue sodium accumulation may contribute to such association. METHODS AND RESULTS: In a cross-sectional study of 30 obese and 53 non-obese subjects, we measured insulin sensitivity determined as glucose disposal rate (GDR) using hyperinsulinemic euglycemic clamp, and tissue sodium content using 23Na magnetic resonance imaging. Median age was 48 years, 68% were female and 41% were African American. Median (interquartile range) BMI was 33 (31.5, 36.3) and 25 (23.5, 27.2) kg/m2 in the obese and non-obese individuals, respectively. In obese individuals, insulin sensitivity negatively correlated with muscle (r = -0.45, p = 0.01) and skin sodium (r = -0.46, p = 0.01). In interaction analysis among obese individuals, tissue sodium had a greater effect on insulin sensitivity at higher levels of high-sensitivity C-reactive protein (p-interaction = 0.03 and 0.01 for muscle and skin Na+, respectively) and interleukin-6 (p-interaction = 0.024 and 0.003 for muscle and skin Na+, respectively). In interaction analysis of the entire cohort, the association between muscle sodium and insulin sensitivity was stronger with increasing levels of serum leptin (p-interaction = 0.01). CONCLUSIONS: Higher muscle and skin sodium are associated with insulin resistance in obese patients. Whether high tissue sodium accumulation has a mechanistic role in the development of obesity-related insulin resistance through systemic inflammation and leptin dysregulation remains to be examined in future studies. CLINICALTRIALS: gov registration: NCT02236520.


Asunto(s)
Resistencia a la Insulina , Humanos , Femenino , Persona de Mediana Edad , Masculino , Leptina , Glucemia/metabolismo , Insulina , Estudios Transversales , Obesidad , Inflamación/diagnóstico , Sodio
5.
JAMA ; 330(23): 2258-2266, 2023 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-37950918

RESUMEN

Importance: Dietary sodium recommendations are debated partly due to variable blood pressure (BP) response to sodium intake. Furthermore, the BP effect of dietary sodium among individuals taking antihypertensive medications is understudied. Objectives: To examine the distribution of within-individual BP response to dietary sodium, the difference in BP between individuals allocated to consume a high- or low-sodium diet first, and whether these varied according to baseline BP and antihypertensive medication use. Design, Setting, and Participants: Prospectively allocated diet order with crossover in community-based participants enrolled between April 2021 and February 2023 in 2 US cities. A total of 213 individuals aged 50 to 75 years, including those with normotension (25%), controlled hypertension (20%), uncontrolled hypertension (31%), and untreated hypertension (25%), attended a baseline visit while consuming their usual diet, then completed 1-week high- and low-sodium diets. Intervention: High-sodium (approximately 2200 mg sodium added daily to usual diet) and low-sodium (approximately 500 mg daily total) diets. Main Outcomes and Measures: Average 24-hour ambulatory systolic and diastolic BP, mean arterial pressure, and pulse pressure. Results: Among the 213 participants who completed both high- and low-sodium diet visits, the median age was 61 years, 65% were female and 64% were Black. While consuming usual, high-sodium, and low-sodium diets, participants' median systolic BP measures were 125, 126, and 119 mm Hg, respectively. The median within-individual change in mean arterial pressure between high- and low-sodium diets was 4 mm Hg (IQR, 0-8 mm Hg; P < .001), which did not significantly differ by hypertension status. Compared with the high-sodium diet, the low-sodium diet induced a decline in mean arterial pressure in 73.4% of individuals. The commonly used threshold of a 5 mm Hg or greater decline in mean arterial pressure between a high-sodium and a low-sodium diet classified 46% of individuals as "salt sensitive." At the end of the first dietary intervention week, the mean systolic BP difference between individuals allocated to a high-sodium vs a low-sodium diet was 8 mm Hg (95% CI, 4-11 mm Hg; P < .001), which was mostly similar across subgroups of age, sex, race, hypertension, baseline BP, diabetes, and body mass index. Adverse events were mild, reported by 9.9% and 8.0% of individuals while consuming the high- and low-sodium diets, respectively. Conclusions and Relevance: Dietary sodium reduction significantly lowered BP in the majority of middle-aged to elderly adults. The decline in BP from a high- to low-sodium diet was independent of hypertension status and antihypertensive medication use, was generally consistent across subgroups, and did not result in excess adverse events. Trial Registration: ClinicalTrials.gov Identifier: NCT04258332.


Asunto(s)
Presión Sanguínea , Hipertensión , Sodio en la Dieta , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Antihipertensivos/uso terapéutico , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Estudios Cruzados , Dieta Hiposódica , Hipertensión/tratamiento farmacológico , Hipertensión/etiología , Hipertensión/fisiopatología , Sodio/farmacología , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio Dietético/farmacología , Sodio en la Dieta/efectos adversos , Sodio en la Dieta/farmacología
6.
Arterioscler Thromb Vasc Biol ; 41(4): 1459-1473, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33567869
7.
FASEB J ; 34(12): 15946-15960, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33015868

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the global pandemic of coronavirus disease-2019 (COVID-19). SARS-CoV-2 is a zoonotic disease, but little is known about variations in species susceptibility that could identify potential reservoir species, animal models, and the risk to pets, wildlife, and livestock. Certain species, such as domestic cats and tigers, are susceptible to SARS-CoV-2 infection, while other species such as mice and chickens are not. Most animal species, including those in close contact with humans, have unknown susceptibility. Hence, methods to predict the infection risk of animal species are urgently needed. SARS-CoV-2 spike protein binding to angiotensin-converting enzyme 2 (ACE2) is critical for viral cell entry and infection. Here we integrate species differences in susceptibility with multiple in-depth structural analyses to identify key ACE2 amino acid positions including 30, 83, 90, 322, and 354 that distinguish susceptible from resistant species. Using differences in these residues across species, we developed a susceptibility score that predicts an elevated risk of SARS-CoV-2 infection for multiple species including horses and camels. We also demonstrate that SARS-CoV-2 is nearly optimal for binding ACE2 of humans compared to other animals, which may underlie the highly contagious transmissibility of this virus among humans. Taken together, our findings define potential ACE2 and SARS-CoV-2 residues for therapeutic targeting and identification of animal species on which to focus research and protection measures for environmental and public health.


Asunto(s)
Enzima Convertidora de Angiotensina 2/química , COVID-19/genética , Predisposición Genética a la Enfermedad , Receptores Virales/química , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2/genética , Animales , Camelus , Glicosilación , Caballos , Humanos , Modelos Moleculares , Filogenia , Unión Proteica , Estructura Terciaria de Proteína , Receptores Virales/genética , SARS-CoV-2 , Alineación de Secuencia , Especificidad de la Especie
8.
Curr Hypertens Rep ; 23(3): 13, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33666761

RESUMEN

PURPOSE OF REVIEW: To summarize key advances in our understanding of the role of interleukin 17A (IL-17A) in the pathogenesis of hypertension and highlight important areas for future research and clinical translation. RECENT FINDINGS: While T helper 17 (Th17) cells are major producers of IL-17A, there are several additional innate and adaptive immune cell sources including gamma-delta T cells, innate lymphoid cells, and natural killer cells. IL-17A promotes an increase in blood pressure through multiple mechanisms including inhibiting endothelial nitric oxide production, increasing reactive oxygen species formation, promoting vascular fibrosis, and enhancing renal sodium retention and glomerular injury. IL-17A production from Th17 cells is increased by high salt conditions in vitro and in vivo. There is also emerging data linking salt, the gut microbiome, and intestinal T cell IL-17A production. Novel therapeutics targeting IL-17A signaling are approved for the treatment of autoimmune diseases and show promise in both animal models of hypertension and human studies. Hypertensive stimuli enhance IL-17A production. IL-17A is a key mediator of renal and vascular dysfunction in hypertensive mouse models and correlates with hypertension in humans. Large randomized clinical trials are needed to determine whether targeting IL-17A might be an effective adjunct treatment for hypertension and its associated end-organ dysfunction.


Asunto(s)
Hipertensión , Interleucina-17 , Animales , Presión Sanguínea , Humanos , Hipertensión/tratamiento farmacológico , Inmunidad Innata , Linfocitos
9.
Arterioscler Thromb Vasc Biol ; 40(6): e153-e165, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32295422

RESUMEN

OBJECTIVE: Macrophages have been described in calcific aortic valve disease, but it is unclear if they promote or counteract calcification. We aimed to determine how macrophages are involved in calcification using the Notch1+/- model of calcific aortic valve disease. Approach and Results: Macrophages in wild-type and Notch1+/- murine aortic valves were characterized by flow cytometry. Macrophages in Notch1+/- aortic valves had increased expression of MHCII (major histocompatibility complex II). We then used bone marrow transplants to test if differences in Notch1+/- macrophages drive disease. Notch1+/- mice had increased valve thickness, macrophage infiltration, and proinflammatory macrophage maturation regardless of transplanted bone marrow genotype. In vitro approaches confirm that Notch1+/- aortic valve cells promote macrophage invasion as quantified by migration index and proinflammatory phenotypes as quantified by Ly6C and CCR2 positivity independent of macrophage genotype. Finally, we found that macrophage interaction with aortic valve cells promotes osteogenic, but not dystrophic, calcification and decreases abundance of the STAT3ß isoform. CONCLUSIONS: This study reveals that Notch1+/- aortic valve disease involves increased macrophage recruitment and maturation driven by altered aortic valve cell secretion, and that increased macrophage recruitment promotes osteogenic calcification and alters STAT3 splicing. Further investigation of STAT3 and macrophage-driven inflammation as therapeutic targets in calcific aortic valve disease is warranted.


Asunto(s)
Estenosis de la Válvula Aórtica/patología , Válvula Aórtica/patología , Calcinosis/patología , Macrófagos/fisiología , Factor de Transcripción STAT3/fisiología , Animales , Válvula Aórtica/inmunología , Válvula Aórtica/fisiopatología , Estenosis de la Válvula Aórtica/inmunología , Estenosis de la Válvula Aórtica/fisiopatología , Trasplante de Médula Ósea , Calcinosis/inmunología , Calcinosis/fisiopatología , Movimiento Celular , Óxidos S-Cíclicos/farmacología , Modelos Animales de Enfermedad , Expresión Génica , Genotipo , Humanos , Inflamación/patología , Macrófagos/química , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteogénesis , Receptor Notch1/análisis , Receptor Notch1/genética , Receptor Notch1/fisiología , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética
10.
Am J Physiol Cell Physiol ; 319(4): C757-C770, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32845718

RESUMEN

Extracellular fluid (ECF) potassium concentration ([K+]) is maintained by adaptations of kidney and skeletal muscle, responses heretofore studied separately. We aimed to determine how these organ systems work in concert to preserve ECF [K+] in male C57BL/6J mice fed a K+-deficient diet (0K) versus 1% K+ diet (1K) for 10 days (n = 5-6/group). During 0K feeding, plasma [K+] fell from 4.5 to 2 mM; hindlimb muscle (gastrocnemius and soleus) lost 28 mM K+ (from 115 ± 2 to 87 ± 2 mM) and gained 27 mM Na+ (from 27 ± 0.4 to 54 ± 2 mM). Doubling of muscle tissue [Na+] was not associated with inflammation, cytokine production or hypertension as reported by others. Muscle transporter adaptations in 0K- versus 1K-fed mice, assessed by immunoblot, included decreased sodium pump α2-ß2 subunits, decreased K+-Cl- cotransporter isoform 3, and increased phosphorylated (p) Na+,K+,2Cl- cotransporter isoform 1 (NKCC1p), Ste20/SPS-1-related proline-alanine rich kinase (SPAKp), and oxidative stress-responsive kinase 1 (OSR1p) consistent with intracellular fluid (ICF) K+ loss and Na+ gain. Renal transporters' adaptations, effecting a 98% reduction in K+ excretion, included two- to threefold increased phosphorylated Na+-Cl- cotransporter (NCCp), SPAKp, and OSR1p abundance, limiting Na+ delivery to epithelial Na+ channels where Na+ reabsorption drives K+ secretion; and renal K sensor Kir 4.1 abundance fell 25%. Mass balance estimations indicate that over 10 days of 0K feeding, mice lose ~48 µmol K+ into the urine and muscle shifts ~47 µmol K+ from ICF to ECF, illustrating the importance of the concerted responses during K+ deficiency.


Asunto(s)
Adaptación Fisiológica/genética , Hipertensión/genética , Riñón/metabolismo , Potasio/metabolismo , Animales , Presión Sanguínea/genética , Canales Epiteliales de Sodio/genética , Líquido Extracelular/metabolismo , Humanos , Hipertensión/patología , Riñón/patología , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Fosforilación/genética , Canales de Potasio de Rectificación Interna/genética , Proteínas Serina-Treonina Quinasas/genética , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Simportadores/genética , Factores de Transcripción/genética , Cotransportadores de K Cl
11.
Am J Physiol Heart Circ Physiol ; 317(1): H141-H155, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31050556

RESUMEN

Calcific aortic valve disease (CAVD) is highly prevalent and has no pharmaceutical treatment. Surgical replacement of the aortic valve has proved effective in advanced disease but is costly, time limited, and in many cases not optimal for elderly patients. This has driven an increasing interest in noninvasive therapies for patients with CAVD. Adaptive immune cell signaling in the aortic valve has shown potential as a target for such a therapy. Up to 15% of cells in the healthy aortic valve are hematopoietic in origin, and these cells, which include macrophages, T lymphocytes, and B lymphocytes, are increased further in calcified specimens. Additionally, cytokine signaling has been shown to play a causative role in aortic valve calcification both in vitro and in vivo. This review summarizes the physiological presence of hematopoietic cells in the valve, innate and adaptive immune cell infiltration in disease states, and the cytokine signaling pathways that play a significant role in CAVD pathophysiology and may prove to be pharmaceutical targets for this disease in the near future.


Asunto(s)
Inmunidad Adaptativa , Estenosis de la Válvula Aórtica/inmunología , Válvula Aórtica/inmunología , Válvula Aórtica/patología , Calcinosis/inmunología , Linfocitos/inmunología , Células Mieloides/inmunología , Animales , Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/patología , Calcinosis/metabolismo , Calcinosis/patología , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Inmunidad Innata , Linfocitos/metabolismo , Linfocitos/patología , Células Mieloides/metabolismo , Células Mieloides/patología , Transducción de Señal
12.
Circ Res ; 118(8): 1233-43, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26988069

RESUMEN

RATIONALE: Accumulating evidence supports a role of adaptive immunity and particularly T cells in the pathogenesis of hypertension. Formation of memory T cells, which requires the costimulatory molecule CD70 on antigen-presenting cells, is a cardinal feature of adaptive immunity. OBJECTIVE: To test the hypothesis that CD70 and immunologic memory contribute to the blood pressure elevation and renal dysfunction mediated by repeated hypertensive challenges. METHODS AND RESULTS: We imposed repeated hypertensive challenges using either N(ω)-nitro-L-arginine methyl ester hydrochloride (L-NAME)/high salt or repeated angiotensin II stimulation in mice. During these challenges effector memory T cells (T(EM)) accumulated in the kidney and bone marrow. In the L-NAME/high-salt model, memory T cells of the kidney were predominant sources of interferon-γ and interleukin-17A, known to contribute to hypertension. L-NAME/high salt increased macrophage and dendritic cell surface expression of CD70 by 3- to 5-fold. Mice lacking CD70 did not accumulate T(EM) cells and did not develop hypertension to either high salt or the second angiotensin II challenge and were protected against renal damage. Bone marrow-residing T(EM) cells proliferated and redistributed to the kidney in response to repeated salt feeding. Adoptively transferred T(EM) cells from hypertensive mice homed to the bone marrow and spleen and expanded on salt feeding of the recipient mice. CONCLUSIONS: Our findings illustrate a previously undefined role of CD70 and long-lived T(EM) cells in the development of blood pressure elevation and end-organ damage that occur on delayed exposure to mild hypertensive stimuli. Interventions to prevent repeated hypertensive surges could attenuate formation of hypertension-specific T(EM) cells.


Asunto(s)
Presión Sanguínea/fisiología , Ligando CD27/deficiencia , Hipertensión/metabolismo , Enfermedades Renales/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Animales , Presión Sanguínea/efectos de los fármacos , Hipertensión/inducido químicamente , Mediadores de Inflamación/metabolismo , Enfermedades Renales/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NG-Nitroarginina Metil Éster/toxicidad , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
13.
Am J Physiol Renal Physiol ; 313(2): F141-F144, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28404590

RESUMEN

Hypertension is growing in epidemic proportions worldwide and is now the leading preventable cause of premature death. For over a century, we have known that the kidney plays a critical role in blood pressure regulation. Specifically, abnormalities in renal sodium transport appear to be a final common pathway that gives rise to elevated blood pressure regardless of the nature of the initial hypertensive stimulus. However, it is only in the past decade that we have come to realize that inflammatory cytokines secreted by innate and adaptive immune cells, as well as renal epithelial cells, can modulate the expression and activity of sodium transporters all along the nephron, leading to alterations in pressure natriuresis, sodium and water balance, and ultimately hypertension. This mini-review highlights specific cytokines and the transporters that they regulate and discusses why inflammatory cytokines may have evolved to serve this function.


Asunto(s)
Presión Sanguínea , Citocinas/metabolismo , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Hipertensión/metabolismo , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Animales , Citocinas/inmunología , Células Epiteliales/inmunología , Canales Epiteliales de Sodio/inmunología , Humanos , Hipertensión/inmunología , Hipertensión/fisiopatología , Mediadores de Inflamación/inmunología , Riñón/inmunología , Riñón/fisiopatología , Proteínas de Transporte de Membrana/inmunología , Reabsorción Renal , Transducción de Señal
14.
Circ Res ; 116(6): 1022-33, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25767287

RESUMEN

For >50 years, it has been recognized that immunity contributes to hypertension. Recent data have defined an important role of T cells and various T cell-derived cytokines in several models of experimental hypertension. These studies have shown that stimuli like angiotensin II, deoxycorticosterone acetate-salt, and excessive catecholamines lead to formation of effector like T cells that infiltrate the kidney and perivascular regions of both large arteries and arterioles. There is also accumulation of monocyte/macrophages in these regions. Cytokines released from these cells, including interleukin-17, interferon-γ, tumor necrosis factorα, and interleukin-6 promote both renal and vascular dysfunction and damage, leading to enhanced sodium retention and increased systemic vascular resistance. The renal effects of these cytokines remain to be fully defined, but include enhanced formation of angiotensinogen, increased sodium reabsorption, and increased renal fibrosis. Recent experiments have defined a link between oxidative stress and immune activation in hypertension. These have shown that hypertension is associated with formation of reactive oxygen species in dendritic cells that lead to formation of gamma ketoaldehydes, or isoketals. These rapidly adduct to protein lysines and are presented by dendritic cells as neoantigens that activate T cells and promote hypertension. Thus, cells of both the innate and adaptive immune system contribute to end-organ damage and dysfunction in hypertension. Therapeutic interventions to reduce activation of these cells may prove beneficial in reducing end-organ damage and preventing consequences of hypertension, including myocardial infarction, heart failure, renal failure, and stroke.


Asunto(s)
Hipertensión/inmunología , Inflamación/inmunología , Inmunidad Adaptativa/fisiología , Animales , Bencilaminas/farmacología , Bencilaminas/uso terapéutico , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/prevención & control , Citocinas/deficiencia , Citocinas/fisiología , Evaluación Preclínica de Medicamentos , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/patología , Hipertensión/fisiopatología , Inmunidad Innata/fisiología , Inflamación/fisiopatología , Riñón/inmunología , Riñón/patología , Riñón/fisiopatología , Activación de Linfocitos , Ratones , Ratones Noqueados , Modelos Animales , Modelos Cardiovasculares , Modelos Inmunológicos , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/fisiología , Remodelación Vascular , Rigidez Vascular
15.
Mol Syst Biol ; 11(1): 799, 2015 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-25882670

RESUMEN

Genome-wide association studies (GWAS) have identified numerous loci associated with blood pressure (BP). The molecular mechanisms underlying BP regulation, however, remain unclear. We investigated BP-associated molecular mechanisms by integrating BP GWAS with whole blood mRNA expression profiles in 3,679 individuals, using network approaches. BP transcriptomic signatures at the single-gene and the coexpression network module levels were identified. Four coexpression modules were identified as potentially causal based on genetic inference because expression-related SNPs for their corresponding genes demonstrated enrichment for BP GWAS signals. Genes from the four modules were further projected onto predefined molecular interaction networks, revealing key drivers. Gene subnetworks entailing molecular interactions between key drivers and BP-related genes were uncovered. As proof-of-concept, we validated SH2B3, one of the top key drivers, using Sh2b3(-/-) mice. We found that a significant number of genes predicted to be regulated by SH2B3 in gene networks are perturbed in Sh2b3(-/-) mice, which demonstrate an exaggerated pressor response to angiotensin II infusion. Our findings may help to identify novel targets for the prevention or treatment of hypertension.


Asunto(s)
Presión Sanguínea/genética , Hipertensión/genética , Proteínas Adaptadoras Transductoras de Señales , Adulto , Anciano , Angiotensina II/metabolismo , Animales , Índice de Masa Corporal , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Redes Reguladoras de Genes , Sitios Genéticos , Estudio de Asociación del Genoma Completo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Lineales , Masculino , Proteínas de la Membrana , Ratones , Ratones Noqueados , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Dominios y Motivos de Interacción de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Biología de Sistemas , Transcriptoma , Adulto Joven
16.
Curr Opin Nephrol Hypertens ; 25(2): 87-93, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26717315

RESUMEN

PURPOSE OF REVIEW: Hypertension is a leading cause of cardiovascular and renal morbidity, and mortality. Genome-wide association studies identified a single-nucleotide polymorphism in the gene SH2B3 encoding the lymphocyte adaptor protein, LNK, but, until recently, little was known about how LNK contributes to hypertension. This review summarizes recent work highlighting a central role for LNK in inflammation and hypertension. RECENT FINDINGS: Using a systems biology approach that integrates genomic data with whole blood transcriptomic data and network modeling, LNK/SH2B3 was identified as a key driver gene for hypertension in humans. LNK is an intracellular adaptor protein expressed predominantly in hematopoietic and endothelial cells that negatively regulates cell proliferation and cytokine signaling. Genetic animal models with deletion or mutation of LNK revealed an important role for LNK in renal and vascular inflammation, glomerular injury, oxidative stress, interferon-γ production, and hypertension. Bone marrow transplantation experiments revealed that LNK in hematopoietic cells is primarily responsible for blood pressure regulation. SUMMARY: LNK/SH2B3 is a key driver gene for human hypertension, and alteration of LNK in animal models has a profound effect on inflammation and hypertension. Thus, LNK is a potential therapeutic target for this disease and its devastating consequences.


Asunto(s)
Hipertensión/genética , Inflamación/genética , Interferón gamma/metabolismo , Proteínas/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Presión Sanguínea , Células Dendríticas/metabolismo , Humanos , Hipertensión/complicaciones , Hipertensión/metabolismo , Inflamación/complicaciones , Inflamación/metabolismo , Interferón gamma/biosíntesis , Péptidos y Proteínas de Señalización Intracelular , Riñón/fisiopatología , Proteínas/metabolismo
17.
Circ Res ; 114(4): 616-25, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24347665

RESUMEN

RATIONALE: Aortic stiffening commonly occurs in hypertension and further elevates systolic pressure. Hypertension is also associated with vascular inflammation and increased mechanical stretch. The interplay between inflammation, mechanical stretch, and aortic stiffening in hypertension remains undefined. OBJECTIVE: Our aim was to determine the role of inflammation and mechanical stretch in aortic stiffening. METHODS AND RESULTS: Chronic angiotensin II infusion caused marked aortic adventitial collagen deposition, as quantified by Masson trichrome blue staining and biochemically by hydroxyproline content, in wild-type but not in recombination activating gene-1-deficient mice. Aortic compliance, defined by ex vivo measurements of stress-strain curves, was reduced by chronic angiotensin II infusion in wild-type mice (P<0.01) but not in recombination activating gene-1-deficient mice (P<0.05). Adoptive transfer of T-cells to recombination activating gene-1-deficient mice restored aortic collagen deposition and stiffness to values observed in wild-type mice. Mice lacking the T-cell-derived cytokine interleukin 17a were also protected against aortic stiffening. In additional studies, we found that blood pressure normalization by treatment with hydralazine and hydrochlorothiazide prevented angiotensin II-induced vascular T-cell infiltration, aortic stiffening, and collagen deposition. Finally, we found that mechanical stretch induces the expression of collagen 1α1, 3α1, and 5a1 in cultured aortic fibroblasts in a p38 mitogen-activated protein kinase-dependent fashion, and that inhibition of p38 prevented angiotensin II-induced aortic stiffening in vivo. Interleukin 17a also induced collagen 3a1 expression via the activation of p38 mitogen-activated protein kinase. CONCLUSIONS: Our data define a pathway in which inflammation and mechanical stretch lead to vascular inflammation that promotes collagen deposition. The resultant increase in aortic stiffness likely further worsens systolic hypertension and its attendant end-organ damage.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Hipertensión/metabolismo , Inflamación/metabolismo , Rigidez Vascular/fisiología , Vasculitis/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Traslado Adoptivo , Angiotensina II/farmacología , Animales , Enfermedades de la Aorta/fisiopatología , Antígenos CD4/genética , Antígenos CD8/genética , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Elastina/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Proteínas de Homeodominio/genética , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Interleucina-17/genética , Masculino , Ratones , Ratones Noqueados , Estrés Mecánico , Linfocitos T/metabolismo , Linfocitos T/patología , Vasculitis/fisiopatología , Vasoconstrictores/farmacología
18.
BMC Nephrol ; 17: 24, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26936597

RESUMEN

BACKGROUND: Two-dimensional measures of vascular architecture provide incomplete information about vascular structure. This study applied a novel rigorous method for 3D microCT-based analysis of total and cortical renal vasculature combined with a novel method to isolate and quantify the number of perfused glomeruli to assess vascular changes in eNOS-/- mice. METHODS: Two month old male wildtype and eNOS-/- mice were perfused with heparinized saline followed by radiopaque Microfil. The Microfil-perfused vasculature of excised kidneys was imaged by µCT with an isotropic voxel-size of 5.0 µm. For analysis of renal cortical vasculature, a custom algorithm was created to define the cortical volume of interest (VOI) as the entire volume within 600 µm of the renal surface. Vessel thickness in the whole kidney or renal cortex was analyzed by plotting the distribution of vascular volume at each measured thickness and examining differences between the genotypes at individual thicknesses. A second image processing algorithm was created to isolate, identify, and extract contrast perfused glomeruli from the cortical vessels. RESULTS: Fractional vascular volume (vascular volume/kidney volume; VV/KV) and Vessel Number/mm (V.N) were significantly lower in eNOS-/- mice vs. WT (p < 0.05). eNOS-/- kidneys had significantly fewer perfusable vessels vs. WT in the range of 20-40 µm in thickness. The cortex of eNOS-/- kidneys had significantly lower VV, VV/cortical volume, and V.N, with an increase in the distance between vessels (all p < 0.05). The total volume of vessels in the range of 20-30 µm was significantly lower in the cortex of eNOS-/- mice compared to WT (p < 0.05). Moreover, the total number of perfused glomeruli was significantly decreased in eNOS-/- mice (p < 0.01). CONCLUSIONS: The methods presented here demonstrate a new method to analyze contrast enhanced µCT images for vascular phenotyping of the murine kidney. These data also demonstrate that kidneys in eNOS-/- mice have severe defects in vascular perfusion/structure in the renal cortex.


Asunto(s)
Arteriolas/diagnóstico por imagen , Corteza Renal/diagnóstico por imagen , Glomérulos Renales/diagnóstico por imagen , Óxido Nítrico Sintasa de Tipo III/genética , Arteria Renal/diagnóstico por imagen , Animales , Corteza Renal/irrigación sanguínea , Glomérulos Renales/irrigación sanguínea , Masculino , Ratones , Ratones Noqueados , Tamaño de los Órganos , Microtomografía por Rayos X/métodos
19.
Antioxid Redox Signal ; 40(4-6): 292-316, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37125445

RESUMEN

Significance: Chronic inflammation has emerged as a major underlying cause of many prevalent conditions in the Western world, including cardiovascular diseases. Although targeting inflammation has emerged as a promising avenue by which to treat cardiovascular disease, it is also associated with increased risk of infection. Recent Advances: Though previously assumed to be passive, resolution has now been identified as an active process, mediated by unique immunoresolving mediators and mechanisms designed to terminate acute inflammation and promote tissue repair. Recent work has determined that failures of resolution contribute to chronic inflammation and the progression of human disease. Specifically, failure to produce pro-resolving mediators and the impaired clearance of dead cells from inflamed tissue have been identified as major mechanisms by which resolution fails in disease. Critical Issues: Drawing from a rapidly expanding body of experimental and clinical studies, we review here what is known about the role of inflammation resolution in arterial hypertension, atherosclerosis, myocardial infarction, and ischemic heart disease. For each, we discuss the involvement of specialized pro-resolving mediators and pro-reparative cell types, including T regulatory cells, myeloid-derived suppressor cells, and macrophages. Future Directions: Pro-resolving therapies offer the promise of limiting chronic inflammation without impairing host defense. Therefore, it is imperative to better understand the mechanisms underlying resolution to identify therapeutic targets. Antioxid. Redox Signal. 40, 292-316.


Asunto(s)
Aterosclerosis , Sistema Cardiovascular , Hipertensión , Infarto del Miocardio , Humanos , Aterosclerosis/metabolismo , Inflamación/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Sistema Cardiovascular/metabolismo , Hipertensión/tratamiento farmacológico , Mediadores de Inflamación/metabolismo
20.
Physiol Rep ; 12(13): e16127, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38960895

RESUMEN

BACKGROUND AND AIMS: High tissue sodium accumulation and intermuscular adipose tissue (IMAT) are associated with aging, type 2 diabetes, and chronic kidney disease. In this study, we aim to investigate whether high lower-extremity tissue sodium accumulation relates to IMAT quantity and whether systemic inflammatory mediators and adipocytokines contribute to such association. METHODS: Tissue sodium content and IMAT accumulation (percentage of IMAT area to muscle area) were measured in 83 healthy individuals using sodium imaging (23Na-MRI) and proton (1H-MRI) imaging of the calf. Insulin sensitivity was assessed by glucose disposal rate (GDR) measured with the hyperinsulinemic-euglycemic clamp. RESULTS: Median (interquartile range) muscle and skin sodium contents were 16.6 (14.9, 19.0) and 12.6 (10.9, 16.7) mmol/L, respectively. Median IMAT was 3.69 (2.80, 5.37) %. In models adjusted for age, sex, BMI, GDR, adiponectin, and high-sensitivity C-reactive protein, increasing tissue sodium content was significantly associated with higher IMAT quantity (p = 0.018 and 0.032 for muscle and skin tissue sodium, respectively). In subgroup analysis stratified by sex, skin sodium was significantly associated with IMAT only among men. In interaction analysis, the association between skin sodium and IMAT was greater with increasing levels of high-sensitivity C-reactive protein and interleukin-6 (p for interaction = 0.022 and 0.006, respectively). CONCLUSIONS: Leg muscle and skin sodium are associated with IMAT quantity among healthy individuals. The relationship between skin sodium and IMAT may be mediated by systemic inflammation.


Asunto(s)
Tejido Adiposo , Músculo Esquelético , Sodio , Humanos , Masculino , Femenino , Tejido Adiposo/metabolismo , Tejido Adiposo/diagnóstico por imagen , Adulto , Sodio/metabolismo , Músculo Esquelético/metabolismo , Persona de Mediana Edad , Piel/metabolismo , Resistencia a la Insulina , Imagen por Resonancia Magnética/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA