Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.563
Filtrar
1.
Circ Res ; 134(11): 1546-1565, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38781300

RESUMEN

Cardiac abnormalities were identified early in the epidemic of AIDS, predating the isolation and characterization of the etiologic agent, HIV. Several decades later, the causation and pathogenesis of cardiovascular disease (CVD) linked to HIV infection continue to be the focus of intense speculation. Before the widespread use of antiretroviral therapy, HIV-associated CVD was primarily characterized by HIV-associated cardiomyopathy linked to profound immunodeficiency. With increasing antiretroviral therapy use, viral load suppression, and establishment of immune competency, the effects of HIV on the cardiovascular system are more subtle. Yet, people living with HIV still face an increased incidence of cardiovascular pathology. Advances in cardiac imaging modalities and immunology have deepened our understanding of the pathogenesis of HIV-associated CVD. This review provides an overview of the pathogenesis of HIV-associated CVD integrating data from imaging and immunologic studies with particular relevance to the HIV population originating from high-endemic regions, such as sub-Saharan Africa. The review highlights key evidence gaps in the field and suggests future directions for research to better understand the complex HIV-CVD interactions.


Asunto(s)
Enfermedades Cardiovasculares , Infecciones por VIH , Humanos , Infecciones por VIH/inmunología , Infecciones por VIH/epidemiología , Infecciones por VIH/complicaciones , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/diagnóstico por imagen , Animales
2.
Nature ; 629(8010): 174-183, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38693412

RESUMEN

Regular exercise promotes whole-body health and prevents disease, but the underlying molecular mechanisms are incompletely understood1-3. Here, the Molecular Transducers of Physical Activity Consortium4 profiled the temporal transcriptome, proteome, metabolome, lipidome, phosphoproteome, acetylproteome, ubiquitylproteome, epigenome and immunome in whole blood, plasma and 18 solid tissues in male and female Rattus norvegicus over eight weeks of endurance exercise training. The resulting data compendium encompasses 9,466 assays across 19 tissues, 25 molecular platforms and 4 training time points. Thousands of shared and tissue-specific molecular alterations were identified, with sex differences found in multiple tissues. Temporal multi-omic and multi-tissue analyses revealed expansive biological insights into the adaptive responses to endurance training, including widespread regulation of immune, metabolic, stress response and mitochondrial pathways. Many changes were relevant to human health, including non-alcoholic fatty liver disease, inflammatory bowel disease, cardiovascular health and tissue injury and recovery. The data and analyses presented in this study will serve as valuable resources for understanding and exploring the multi-tissue molecular effects of endurance training and are provided in a public repository ( https://motrpac-data.org/ ).


Asunto(s)
Entrenamiento Aeróbico , Multiómica , Condicionamiento Físico Animal , Resistencia Física , Animales , Femenino , Humanos , Masculino , Ratas , Acetilación , Sangre/inmunología , Sangre/metabolismo , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/metabolismo , Bases de Datos Factuales , Epigenoma , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Internet , Lipidómica , Metaboloma , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Especificidad de Órganos/fisiología , Fosforilación , Condicionamiento Físico Animal/fisiología , Resistencia Física/genética , Resistencia Física/fisiología , Proteoma/metabolismo , Proteómica , Factores de Tiempo , Transcriptoma/genética , Ubiquitinación , Heridas y Lesiones/genética , Heridas y Lesiones/inmunología , Heridas y Lesiones/metabolismo
3.
Adv Protein Chem Struct Biol ; 140: 381-417, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38762276

RESUMEN

Cardiovascular disease (CVD) and cancer are major contributors to global morbidity and mortality. This book chapter delves into the intricate relationship between the immune system and the pathogenesis of both cardiovascular and cancer diseases, exploring the roles of innate and adaptive immunities, immune regulation, and immunotherapy in these complex conditions. The innate immune system acts as the first line of defense against tissue damage and infection, with a significant impact on the initiation and progression of CVD and cancer. Endothelial dysfunction, a hallmark in CVD, shares commonalities with the tumor microenvironment in cancer, emphasizing the parallel involvement of the immune system in both conditions. The adaptive immune system, particularly T cells, contributes to prolonged inflammation in both CVD and cancer. Regulatory T cells and the intricate balance between different T cell subtypes influence disease progression, wound healing, and the outcomes of ischemic injury and cancer immunosurveillance. Dysregulation of immune homeostasis can lead to chronic inflammation, contributing to the development and progression of both CVD and cancer. Thus, immunotherapy emerged as a promising avenue for preventing and managing these diseases, with strategies targeting immune cell modulation, cytokine manipulation, immune checkpoint blockade, and tolerance induction. The impact of gut microbiota on CVD and cancer too is explored in this chapter, highlighting the role of gut leakiness, microbial metabolites, and the potential for microbiome-based interventions in cardiovascular and cancer immunotherapies. In conclusion, immunomodulatory strategies and immunotherapy hold promise in reshaping the landscape of cardiovascular and cancer health. Additionally, harnessing the gut microbiota for immune modulation presents a novel approach to prevent and manage these complex diseases, emphasizing the importance of personalized and precision medicine in healthcare. Ongoing research and clinical trials are expected to further elucidate the complex immunological underpinnings of CVD and cancer thereby refining these innovative approaches.


Asunto(s)
Enfermedades Cardiovasculares , Neoplasias , Humanos , Neoplasias/inmunología , Neoplasias/terapia , Enfermedades Cardiovasculares/inmunología , Inmunoterapia , Inmunidad Innata/inmunología , Microbioma Gastrointestinal/inmunología , Animales , Inmunidad Adaptativa/inmunología
4.
Cell Signal ; 119: 111169, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38599440

RESUMEN

Cardiac resident macrophages (CRMs) are essential in maintaining the balance of the immune homeostasis in the heart. One of the main factors in the progression of cardiovascular diseases, such as myocarditis, myocardial infarction(MI), and heart failure(HF), is the imbalance in the regulatory mechanisms of CRMs. Recent studies have reported novel heterogeneity and spatiotemporal complexity of CRMs, and their role in maintaining cardiac immune homeostasis and treating cardiovascular diseases. In this review, we focus on the functions of CRMs, including immune surveillance, immune phagocytosis, and immune metabolism, and explore the impact of CRM's homeostasis imbalance on cardiac injury and cardiac repair. We also discuss the therapeutic approaches linked to CRMs. The immunomodulatory strategies targeting CRMs may be a therapeutic approach for the treatment of cardiovascular disease.


Asunto(s)
Homeostasis , Macrófagos , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Animales , Miocardio/inmunología , Miocardio/metabolismo , Miocardio/patología , Fagocitosis , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/metabolismo
5.
Int J Cardiovasc Imaging ; 40(4): 945-948, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38558330

RESUMEN

In the last century, there has been more than enough research that proved the association of high lipid and glucose levels with cardiovascular disease, thus establishing the current well-known traditional cardiovascular risk factors such as dyslipidemia, diabetes, and metabolic syndrome. Hence, these cardiovascular risk factors are target therapy for glucose and lipid-lowering agents to prevent adverse cardiovascular events. However, despite controlling the lipid and glucose levels, some studies demonstrated the subclinical atherosclerosis suggesting that these cardiovascular risk factors alone cannot account for the entire atherosclerosis burden. In the last years, large-scale clinical trials demonstrated the operation of the inflammatory pathway in atherosclerotic cardiovascular disease (ASCVD) by the immune system, both the innate (neutrophils, macrophages) and adaptive (T cell and other lymphocytes) limbs, contribute to atherosclerosis and atherothrombosis. In this regard, some studies that use antiinflammatory therapy targeting the immune system by modulating or blocking interleukins, also known as anti-cytokine therapy, have been shown to reduce the risk of adverse cardiovascular events in patients with previous coronary artery disease. In this regard, the U.S. Food and Drug Administration (FDA) approved the use of colchicine 0.5 mg once daily for reducing cardiovascular events in patients who have established ASCVD and high residual systemic inflammation. Therefore, measuring the systemic inflammation can improve the cardiovascular risk assessment and identify the subsets of patients that will benefit from anti-cytokine therapy after diagnosis of ASCVD or after myocardial revascularization.


Asunto(s)
Antiinflamatorios , Biomarcadores , Glucemia , Citocinas , Factores de Riesgo de Enfermedad Cardiaca , Mediadores de Inflamación , Inflamación , Triglicéridos , Humanos , Antiinflamatorios/uso terapéutico , Antiinflamatorios/efectos adversos , Aterosclerosis/inmunología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/sangre , Biomarcadores/sangre , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Enfermedades Cardiovasculares/prevención & control , Enfermedades Cardiovasculares/inmunología , Colchicina/uso terapéutico , Colchicina/efectos adversos , Citocinas/sangre , Citocinas/metabolismo , Inflamación/inmunología , Inflamación/tratamiento farmacológico , Inflamación/sangre , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Medición de Riesgo , Factores de Riesgo , Resultado del Tratamiento , Triglicéridos/sangre
6.
Free Radic Biol Med ; 219: 64-75, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38604314

RESUMEN

Cardiovascular diseases (CVDs) are the leading cause of death globally, resulting in a major health burden. Thus, an urgent need exists for exploring effective therapeutic targets to block progression of CVDs and improve patient prognoses. Immune and inflammatory responses are involved in the development of atherosclerosis, ischemic myocardial damage responses and repair, calcification, and stenosis of the aortic valve. These responses can involve both large and small blood vessels throughout the body, leading to increased blood pressure and end-organ damage. While exploring potential avenues for therapeutic intervention in CVDs, researchers have begun to focus on immune metabolism, where metabolic changes that occur in immune cells in response to exogenous or endogenous stimuli can influence immune cell effector responses and local immune signaling. Itaconate, an intermediate metabolite of the tricarboxylic acid (TCA) cycle, is related to pathophysiological processes, including cellular metabolism, oxidative stress, and inflammatory immune responses. The expression of immune response gene 1 (IRG1) is upregulated in activated macrophages, and this gene encodes an enzyme that catalyzes the production of itaconate from the TCA cycle intermediate, cis-aconitate. Itaconate and its derivatives have exerted cardioprotective effects through immune modulation in various disease models, such as ischemic heart disease, valvular heart disease, vascular disease, heart transplantation, and chemotherapy drug-induced cardiotoxicity, implying their therapeutic potential in CVDs. In this review, we delve into the associated signaling pathways through which itaconate exerts immunomodulatory effects, summarize its specific roles in CVDs, and explore emerging immunological therapeutic strategies for managing CVDs.


Asunto(s)
Enfermedades Cardiovasculares , Succinatos , Humanos , Succinatos/metabolismo , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/patología , Ciclo del Ácido Cítrico , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Carboxiliasas
7.
J Cell Physiol ; 239(5): e31229, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38426269

RESUMEN

RNA-binding proteins (RBPs) play a crucial role in the regulation of posttranscriptional RNA networks, which can undergo dysregulation in many pathological conditions. Human antigen R (HuR) is a highly researched RBP that plays a crucial role as a posttranscriptional regulator. HuR plays a crucial role in the amplification of inflammatory signals by stabilizing the messenger RNA of diverse inflammatory mediators and key molecular players. The noteworthy correlations between HuR and its target molecules, coupled with the remarkable impacts reported on the pathogenesis and advancement of multiple diseases, position HuR as a promising candidate for therapeutic intervention in diverse inflammatory conditions. This review article examines the significance of HuR as a member of the RBP family, its regulatory mechanisms, and its implications in the pathophysiology of inflammation and cardiometabolic illnesses. Our objective is to illuminate potential directions for future research and drug development by conducting a comprehensive analysis of the existing body of research on HuR.


Asunto(s)
Enfermedades Cardiovasculares , Proteína 1 Similar a ELAV , Inflamación , Humanos , Proteína 1 Similar a ELAV/metabolismo , Proteína 1 Similar a ELAV/genética , Inflamación/genética , Inflamación/patología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/metabolismo , Animales , Regulación de la Expresión Génica , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/metabolismo , Transducción de Señal , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
8.
Cardiovasc Res ; 120(6): 567-580, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38395029

RESUMEN

Hypertension is a major cause of cardiovascular diseases such as myocardial infarction and stroke. Cardiovascular fibrosis occurs with hypertension and contributes to vascular resistance, aortic stiffness, and cardiac hypertrophy. However, the molecular mechanisms leading to fibroblast activation in hypertension remain largely unknown. There are two types of fibrosis: replacement fibrosis and reactive fibrosis. Replacement fibrosis occurs in response to the loss of viable tissue to form a scar. Reactive fibrosis occurs in response to an increase in mechanical and neurohormonal stress. Although both types of fibrosis are considered adaptive processes, they become maladaptive when the tissue loss is too large, or the stress persists. Myofibroblasts represent a subpopulation of activated fibroblasts that have gained contractile function to promote wound healing. Therefore, myofibroblasts are a critical cell type that promotes replacement fibrosis. Although myofibroblasts were recognized as the fibroblasts participating in reactive fibrosis, recent experimental evidence indicated there are distinct fibroblast populations in cardiovascular reactive fibrosis. Accordingly, we will discuss the updated definition of fibroblast subpopulations, the regulatory mechanisms, and their potential roles in cardiovascular pathophysiology utilizing new knowledge from various lineage tracing and single-cell RNA sequencing studies. Among the fibroblast subpopulations, we will highlight the novel roles of matrifibrocytes and immune fibrocytes in cardiovascular fibrosis including experimental models of hypertension, pressure overload, myocardial infarction, atherosclerosis, aortic aneurysm, and nephrosclerosis. Exploration into the molecular mechanisms involved in the differentiation and activation of those fibroblast subpopulations may lead to novel treatments for end-organ damage associated with hypertension and other cardiovascular diseases.


Asunto(s)
Fibrosis , Hipertensión , Miofibroblastos , Humanos , Miofibroblastos/patología , Miofibroblastos/metabolismo , Animales , Hipertensión/fisiopatología , Hipertensión/metabolismo , Hipertensión/patología , Hipertensión/inmunología , Miocardio/patología , Miocardio/metabolismo , Miocardio/inmunología , Presión Sanguínea , Transducción de Señal , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/inmunología , Fenotipo
9.
J Leukoc Biol ; 112(6): 1649-1661, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36073777

RESUMEN

Due to the ability of γδ T cells to bridge adaptive and innate immunity, γδ T cells can respond to a variety of molecular cues and acquire the ability to induce a variety of cytokines such as IL-17 family, IFN-γ, IL-4, and IL-10. IL-17+ γδ T cells (γδ T17 cells) populations have recently received considerable interest as they are the major early source of IL-17A in many immune response models. However, the exact mechanism of γδ T17 cells is still poorly understood, especially in the context of cardiovascular disease (CVD). CVD is the leading cause of death in the world, and it tends to be younger. Here, we offer a review of the cardiovascular inflammatory and immune functions of γδ T17 cells in order to understand their role in CVD, which may be the key to developing new clinical applications.


Asunto(s)
Enfermedades Cardiovasculares , Células Th17 , Humanos , Enfermedades Cardiovasculares/inmunología , Inmunidad Innata , Interleucina-17 , Receptores de Antígenos de Linfocitos T gamma-delta , Subgrupos de Linfocitos T , Células Th17/inmunología
11.
Front Immunol ; 13: 879600, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35720418

RESUMEN

Background: Transgender women (TW) are at increased risk for both human immunodeficiency virus (HIV) and cardiovascular disease (CVD). Antiretroviral therapy-treated HIV has been associated with a two-fold increased risk of CVD, potentially due to dysregulated Toll-like receptor (TLR)-induced immune activation. Use of estrogens in feminizing hormone therapy (FHT) may enhance inflammatory responses and the risk of cardiovascular mortality in TW. Despite this, the immunomodulatory effects of estrogen use in TW with HIV have been inadequately explored. Methods: As an in vitro model for FHT, cryopreserved PBMCs (cryoPBMCs) from HIV negative (HIV-), HIV+ ART-suppressed (HIV+SP), and HIV+ ART-unsuppressed (HIV+USP) cisgender men were cultured overnight in the presence of 17-ß estradiol or 17-α ethinylestradiol with and without the TLR4 agonist LPS or the TLR8 agonist ssPolyU. Monocyte activation (CD69, HLA-DR, CD38) was assessed by flow cytometry. Cytokine levels (IL-6, TNF-α, IL-1ß, and IL-10) were measured in cell culture supernatants by Legendplex. Levels of phosphorylated TLR signaling molecules (JNK, MAPK p38) were assessed by Phosflow. Plasma levels of immune activation biomarkers (LPS-binding protein, monocyte activation markers sCD14 and sCD163, and inflammatory molecules IL-6 and TNF-α receptor I) were measured by ELISA. Results: PBMCs from people with HIV (PWH) produced greater levels of inflammatory cytokines following exposure to LPS or ssPolyU compared to levels from cells of HIV- individuals. While estrogen exposure alone induced mild changes in immune activation, LPS-induced TLR4 activation was elevated with estrogen in cisgender men (CM) with HIV, increasing monocyte activation and inflammatory cytokine production (IL-6, TNF-α). Interestingly, testosterone inhibited LPS-induced cytokine production in CM regardless of HIV status. Plasma markers of immune activation and microbial translocation (e.g., sCD14, sCD163, LPS-binding protein) were generally higher in PWH compared to HIV- CM, and these markers were positively associated with in vitro responsiveness to estrogen and LPS in CM with HIV. Conclusions: Our in vitro data suggest that estrogen exposure may enhance innate immune activation in PWH. Further examination is needed to fully understand the complex interactions of FHT, HIV, and CVD in TW, and determine optimal FHT regimens or supplementary treatments aimed at reducing excess immune activation.


Asunto(s)
Estrógenos , Infecciones por VIH , Receptor Toll-Like 4 , Personas Transgénero , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/virología , Citocinas/metabolismo , Estrógenos/efectos adversos , Estrógenos/farmacología , Femenino , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Interleucina-6/inmunología , Receptores de Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Masculino , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/inmunología
12.
Circ Res ; 130(4): 593-610, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35175848

RESUMEN

Immune responses differ between men and women, with women at higher risk of developing chronic autoimmune diseases and having more robust immune responses to many viruses, including HIV and hepatitis C virus. Although immune dysregulation plays a prominent role in chronic systemic inflammation, a key driver in the development of atherosclerotic cardiovascular disease (ASCVD), standard ASCVD risk prediction scores underestimate risk in populations with immune disorders, particularly women. This review focuses on the ASCVD implications of immune dysregulation due to disorders with varying global prevalence by sex: autoimmune disorders (female predominant), HIV (male-female equivalent), and hepatitis C virus (male predominant). Factors contributing to ASCVD in women with immune disorders, including traditional risk factors, dysregulated innate and adaptive immunity, sex hormones, and treatment modalities, are discussed. Finally, the need to develop new ASCVD risk stratification tools that incorporate variables specific to populations with chronic immune disorders, particularly in women, is emphasized.


Asunto(s)
Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/inmunología , Hormonas Esteroides Gonadales/inmunología , Enfermedades del Sistema Inmune/epidemiología , Enfermedades del Sistema Inmune/inmunología , Inmunidad Adaptativa/inmunología , Enfermedades Cardiovasculares/diagnóstico , Femenino , Infecciones por VIH/diagnóstico , Infecciones por VIH/epidemiología , Infecciones por VIH/inmunología , Hepatitis C/diagnóstico , Hepatitis C/epidemiología , Hepatitis C/inmunología , Humanos , Enfermedades del Sistema Inmune/diagnóstico
13.
Life Sci ; 294: 120392, 2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-35149115

RESUMEN

The SARS coronavirus 2 (SARS CoV-2) causes Coronavirus Disease (COVID-19), is an emerging viral infection. SARS CoV-2 infects target cells by attaching to Angiotensin-Converting Enzyme (ACE2). SARS CoV-2 could cause cardiac damage in patients with severe COVID-19, as ACE2 is expressed in cardiac cells, including cardiomyocytes, pericytes, and fibroblasts, and coronavirus could directly infect these cells. Cardiovascular disorders are the most frequent comorbidity found in COVID-19 patients. Immune cells such as monocytes, macrophages, and T cells may produce inflammatory cytokines and chemokines that contribute to COVID-19 pathogenesis if their functions are uncontrolled. This causes a cytokine storm in COVID-19 patients, which has been associated with cardiac damage. Tregs are a subset of immune cells that regulate immune and inflammatory responses. Tregs suppress inflammation and improve cardiovascular function through a variety of mechanisms. This is an exciting research area to explore the cellular, molecular, and immunological mechanisms related to reducing risks of cardiovascular complications in severe COVID-19. This review evaluated whether Tregs can affect COVID-19-related cardiovascular complications, as well as the mechanisms through which Tregs act.


Asunto(s)
COVID-19/inmunología , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/prevención & control , SARS-CoV-2 , Linfocitos T Reguladores/fisiología , Traslado Adoptivo , Animales , Enfermedades Cardiovasculares/virología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/prevención & control , Humanos , Inflamación/inmunología , Linfocitos T Reguladores/inmunología
14.
Viruses ; 14(2)2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35215967

RESUMEN

Zika virus (ZIKV), a re-emerging virus, causes congenital brain abnormalities and Guillain-Barré syndrome. It is mainly transmitted by Aedes mosquitoes, but infections are also linked to sexual transmissions. Infectious ZIKV has been isolated, and viral RNA has been detected in semen over a year after the onset of initial symptoms, but the mode of long-term persistence is not yet understood. ZIKV can proliferate in human Sertoli cells (HSerC) for several weeks in vitro, suggesting that it might be a reservoir for persistent ZIKV infection. This study determined proteomic changes in HSerC during ZIKV infections by TMT-mass spectrometry analysis. Levels of 4416 unique Sertoli cell proteins were significantly altered at 3, 5, and 7 days after ZIKV infection. The significantly altered proteins include enzymes, transcription regulators, transporters, kinases, peptidases, transmembrane receptors, cytokines, ion channels, and growth factors. Many of these proteins are involved in pathways associated with antiviral response, antigen presentation, and immune cell activation. Several immune response pathway proteins were significantly activated during infection, e.g., interferon signaling, T cell receptor signaling, IL-8 signaling, and Th1 signaling. The altered protein levels were linked to predicted activation of immune response in HSerC, which was predicted to suppress ZIKV infection. ZIKV infection also affected the levels of critical regulators of gluconeogenesis and glycolysis pathways such as phosphoglycerate mutase, phosphoglycerate kinase, and enolase. Interestingly, many significantly altered proteins were associated with cardiac hypertrophy, which may induce heart failure in infected patients. In summary, our research contributes to a better understanding of ZIKV replication dynamics and infection in Sertoli cells.


Asunto(s)
Semen/virología , Células de Sertoli/inmunología , Replicación Viral , Infección por el Virus Zika/inmunología , Metabolismo de los Hidratos de Carbono/inmunología , Enfermedades Cardiovasculares/inmunología , Transmisión de Enfermedad Infecciosa , Humanos , Masculino , Procesamiento Proteico-Postraduccional , Proteómica , ARN Viral/genética , Células de Sertoli/virología , Virus Zika/aislamiento & purificación , Infección por el Virus Zika/transmisión
15.
J Clin Invest ; 132(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35040437

RESUMEN

Suppressing inflammation has been the primary focus of therapies in autoimmune rheumatic diseases (AIRDs), including rheumatoid arthritis and systemic lupus erythematosus. However, conventional therapies with low target specificity can have effects on cell metabolism that are less predictable. A key example is lipid metabolism; current therapies can improve or exacerbate dyslipidemia. Many conventional drugs also require in vivo metabolism for their conversion into therapeutically beneficial products; however, drug metabolism often involves the additional formation of toxic by-products, and rates of drug metabolism can be heterogeneous between patients. New therapeutic technologies and research have highlighted alternative metabolic pathways that can be more specifically targeted to reduce inflammation but also to prevent undesirable off-target metabolic consequences of conventional antiinflammatory therapies. This Review highlights the role of lipid metabolism in inflammation and in the mechanisms of action of AIRD therapeutics. Opportunities for cotherapies targeting lipid metabolism that could reduce immunometabolic complications and potential increased cardiovascular disease risk in patients with AIRDs are discussed.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Cardiovasculares/inmunología , Dislipidemias/inmunología , Metabolismo de los Lípidos/inmunología , Enfermedades Reumáticas/inmunología , Enfermedades Autoinmunes/terapia , Enfermedades Cardiovasculares/terapia , Dislipidemias/terapia , Humanos , Inflamación/inmunología , Inflamación/terapia , Enfermedades Reumáticas/terapia
16.
Int J Mol Sci ; 23(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35055116

RESUMEN

Angiotensin II receptor type 1 (AT1R) and endothelin-1 receptor type A (ETAR) are G-protein-coupled receptors (GPCRs) expressed on the surface of a great variety of cells: immune cells, vascular smooth cells, endothelial cells, and fibroblasts express ETAR and AT1R, which are activated by endothelin 1 (ET1) and angiotensin II (AngII), respectively. Certain autoantibodies are specific for these receptors and can regulate their function, thus being known as functional autoantibodies. The function of these antibodies is similar to that of natural ligands, and it involves not only vasoconstriction, but also the secretion of proinflammatory cytokines (such as interleukin-6 (IL6), IL8 and TNF-α), collagen production by fibroblasts, and reactive oxygen species (ROS) release by fibroblasts and neutrophils. The role of autoantibodies against AT1R and ETAR (AT1R-AAs and ETAR-AAs, respectively) is well described in the pathogenesis of many medical conditions (e.g., systemic sclerosis (SSc) and SSc-associated pulmonary hypertension, cystic fibrosis, and allograft dysfunction), but their implications in cardiovascular diseases are still unclear. This review summarizes the current evidence regarding the effects of AT1R-AAs and ETAR-AAs in cardiovascular pathologies, highlighting their roles in heart transplantation and mechanical circulatory support, preeclampsia, and acute coronary syndromes.


Asunto(s)
Autoanticuerpos/metabolismo , Enfermedades Cardiovasculares/inmunología , Receptor de Angiotensina Tipo 1/inmunología , Receptor de Endotelina A/inmunología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Colágeno/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
J Clin Endocrinol Metab ; 107(2): e500-e514, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34597384

RESUMEN

CONTEXT: Vascular aging, including endothelial dysfunction secondary to oxidative stress and inflammation, increases the risk for age-associated cardiovascular disease (CVD). Low testosterone in middle-aged/older men is associated with increased CVD risk. OBJECTIVE: We hypothesized that low testosterone contributes to age-associated endothelial dysfunction, related in part to greater oxidative stress and inflammation. METHODS: This cross-sectional study included 58 healthy, nonsmoking men categorized as young (N = 20; age 29 ± 4 years; testosterone 500 ± 58 ng/dL), middle-aged/older with higher testosterone (N = 20; age 60 ± 6 years; testosterone 512 ± 115 ng/dL), and middle-aged/older lower testosterone (N = 18; age 59 ± 8 years; testosterone 269 ± 48 ng/dL). Brachial artery flow-mediated dilation (FMDBA) was measured during acute infusion of saline (control) and vitamin C (antioxidant). Markers of oxidative stress (total antioxidant status and oxidized low-density lipoprotein cholesterol), inflammation (interleukin [IL]-6 and C-reactive protein [CRP]), and androgen deficiency symptoms were also examined. RESULTS: During saline, FMDBA was reduced in middle-aged/older compared with young, regardless of testosterone status (P < 0.001). FMDBA was reduced in middle-aged/older lower testosterone (3.7% ± 2.0%) compared with middle-aged/older higher testosterone (5.7% ± 2.2%; P = 0.021), independent of symptoms. Vitamin C increased FMDBA (to 5.3% ± 1.6%; P = 0.022) in middle-aged/older lower testosterone but had no effect in young (P = 0.992) or middle-aged/older higher testosterone (P = 0.250). FMDBA correlated with serum testosterone (r = 0.45; P < 0.001), IL-6 (r = -0.41; P = 0.002), and CRP (r = -0.28; P = 0.041). CONCLUSION: Healthy middle-aged/older men with low testosterone appear to have greater age-associated endothelial dysfunction, related in part to greater oxidative stress and inflammation. These data suggest that low testosterone concentrations may contribute to accelerated vascular aging in men.


Asunto(s)
Envejecimiento/metabolismo , Enfermedades Cardiovasculares/epidemiología , Endotelio Vascular/fisiopatología , Testosterona/deficiencia , Adolescente , Adulto , Anciano , Envejecimiento/sangre , Envejecimiento/inmunología , Velocidad del Flujo Sanguíneo , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Estudios Transversales , Endotelio Vascular/diagnóstico por imagen , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Masculino , Persona de Mediana Edad , Estrés Oxidativo/inmunología , Pletismografía , Testosterona/sangre , Ultrasonografía Doppler , Adulto Joven
18.
J Clin Pathol ; 75(1): 30-33, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33785545

RESUMEN

AIMS: So far, little has been known on whether myocardial inflammatory infiltration influences heart failure (HF) progression. Thus, the aim of this study was to test the impact of intramyocardial infiltration on clinical outcomes. METHODS: Biopsy samples from 358 patients with stable HF secondary to dilated cardiomyopathy were studied. Immunohistochemistry for lymphocyte (CD3) and macrophage (CD68) markers was performed and counted. After a 1-year follow-up, patients were classified as improved based on the predefined definition of improvement. The clinical data were collected from 324 patients (90.5%). RESULTS: According to the predefined definition of improvement, 133 patients improved (41.0%) but 191 remained unchanged or deteriorated (58.9%). After a 12-month follow-up, the OR with 95% CI of counts of myocardial inflammatory CD68-positive ≥4 cell/high power field (HPF) compared with CD68-positive <4 cell/HPF for lack of improvement was 1.91 (1.65-2.54). However, the number of CD3 positive cell infiltration had no impact on clinical outcome after a 1-year follow-up. In the baseline study, a reasonably negative correlation was found between the number of CD68 positive cells and troponin T (r=-0.39; p<0.001 by Spearman's r). This was corroborated with a low negative correlation between these cells and myocardial form of creatine kinase (CK-MB) fraction (r=-0.27; p=0.006). There was no correlation between CD3 and CD68 positive cells (Spearman's r; r=-0.17, p=0.16). CONCLUSIONS: The current results provide evidence that high macrophage counts may be a predisposing factor for HF progression.


Asunto(s)
Cardiomiopatía Dilatada/diagnóstico , Enfermedades Cardiovasculares/diagnóstico , Insuficiencia Cardíaca/diagnóstico , Adulto , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores/metabolismo , Biopsia , Complejo CD3/metabolismo , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/patología , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/patología , Femenino , Insuficiencia Cardíaca/inmunología , Insuficiencia Cardíaca/patología , Humanos , Inmunohistoquímica , Inflamación , Linfocitos/inmunología , Linfocitos/patología , Macrófagos/inmunología , Macrófagos/patología , Masculino , Persona de Mediana Edad , Miocardio/inmunología , Miocardio/patología , Pronóstico
19.
J Allergy Clin Immunol ; 149(2): 550-556.e2, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34800432

RESUMEN

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 Enfermedad
20.
J Cell Mol Med ; 26(2): 274-286, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34894069

RESUMEN

Based on the recent reports, cardiovascular events encompass a large portion of the mortality caused by the COVID-19 pandemic, which drawn cardiologists into the management of the admitted ill patients. Given that common laboratory values may provide key insights into the illness caused by the life-threatening SARS-CoV-2 virus, it would be more helpful for screening, clinical management and on-time therapeutic strategies. Commensurate with these issues, this review article aimed to discuss the dynamic changes of the common laboratory parameters during COVID-19 and their association with cardiovascular diseases. Besides, the values that changed in the early stage of the disease were considered and monitored during the recovery process. The time required for returning biomarkers to basal levels was also discussed. Finally, of particular interest, we tended to abridge the latest updates regarding the cardiovascular biomarkers as prognostic and diagnostic criteria to determine the severity of COVID-19.


Asunto(s)
COVID-19/sangre , Enfermedades Cardiovasculares/sangre , Sistema Cardiovascular/metabolismo , SARS-CoV-2/patogenicidad , Biomarcadores/sangre , COVID-19/complicaciones , COVID-19/diagnóstico , COVID-19/inmunología , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/inmunología , Sistema Cardiovascular/patología , Sistema Cardiovascular/virología , Quimiocina CCL2/sangre , Forma MB de la Creatina-Quinasa/sangre , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Homocisteína/sangre , Humanos , Interferón gamma/sangre , Interleucina-6/sangre , L-Lactato Deshidrogenasa/sangre , Péptido Natriurético Encefálico/sangre , Fragmentos de Péptidos/sangre , Pronóstico , SARS-CoV-2/crecimiento & desarrollo , SARS-CoV-2/inmunología , Troponina I/sangre , Troponina T/sangre , Factor de Necrosis Tumoral alfa/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA