Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.592
Filter
1.
Sci Rep ; 14(1): 15129, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956376

ABSTRACT

The systemic immune-inflammation index (SII), a metric reflecting systemic inflammatory response and immune activation, remains underexplored concerning its correlation with mortality among rheumatoid arthritis (RA) patients. This study aimed to delineate the association between SII and both all-cause and cardiovascular mortality within the cohort of American adults diagnosed with RA, utilizing data from the National Health and Nutrition Examination Survey (NHANES) spanning 1999 to 2018. The investigation extracted data from NHANES cycles between 1999 and 2018, identifying RA patients through questionnaire responses. The SII was computed based on complete blood counts, employing the formula: (platelets × neutrophils) / lymphocytes. The optimal SII cutoff value for significant survival outcomes was determined using maximally selected rank statistics. Multivariable Cox proportional hazards models assessed the relationship between SII levels and mortality (all-cause and cardiovascular) among RA patients, with subgroup analyses examining potential modifications by clinical confounders. Additionally, restricted cubic spline (RCS) analyses were conducted to explore the linearity of the SII-mortality association. The study encompassed 2070 American adults with RA, among whom 287 exhibited a higher SII (≥ 919.75) and 1783 a lower SII (< 919.75). Over a median follow-up duration of 108 months, 602 participants died. After adjustments for demographic, socioeconomic, and lifestyle variables, a higher SII was associated with a 1.48-fold increased risk of all-cause mortality (hazard ratio [HR] = 1.48, 95% confidence interval [CI] 1.21-1.81, P < 0.001) and a 1.51-fold increased risk of cardiovascular mortality (HR = 1.51, 95% CI 1.04-2.18, P = 0.030) compared to a lower SII. Kaplan-Meier analyses corroborated significantly reduced survival rates within the higher SII cohort for both all-cause and cardiovascular mortality (Pall-cause mortality < 0.0001 and Pcardiovascular mortality = 0.0004). RCS analyses confirmed a positive nonlinear relationship between SII and mortality rates. In conclusion, the SII offers a straightforward indicator of the equilibrium between detrimental innate inflammation and beneficial adaptive immunity. Our investigation, utilizing a comprehensive and nationally representative sample, reveals that elevated SII levels independently forecast a greater risk of mortality from all causes, as well as cardiovascular-specific mortality, in individuals suffering from RA. These insights underscore the clinical relevance of the SII as an affordable and readily accessible biomarker. Its incorporation into regular clinical practice could significantly enhance the precision of risk assessment and forecasting for patients with RA, facilitating more tailored and effective management strategies. Specifically, patients with high SII levels could be identified for more stringent cardiovascular risk management, including closer monitoring, lifestyle interventions, and aggressive pharmacological treatments to mitigate their increased risk of mortality.


Subject(s)
Arthritis, Rheumatoid , Cardiovascular Diseases , Inflammation , Nutrition Surveys , Humans , Arthritis, Rheumatoid/mortality , Arthritis, Rheumatoid/immunology , Male , Female , Cardiovascular Diseases/mortality , Cardiovascular Diseases/immunology , Middle Aged , Inflammation/immunology , Aged , Adult , Cause of Death , Proportional Hazards Models , Risk Factors
2.
Sci Rep ; 14(1): 16639, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026013

ABSTRACT

Rheumatoid arthritis (RA) is a chronic systemic inflammatory autoimmune disease. However, the relationship between the systemic immune-inflammation index (SII) and the prognosis of RA patients remains unclear. This study aimed to investigate the association between inflammatory biomarker SII and all-cause and cardiovascular mortality in RA patients. A retrospective analysis was conducted using data from the National Health and Nutrition Examination Survey database spanning from 1999 to March 2020. We assessed the association between the SII and all-cause as well as cardiovascular mortality in RA patients employing multivariable Cox proportional hazards regression analysis and restricted cubic spline plots. Receiver operating characteristic curves were employed to evaluate the prognostic capacity of SII in predicting outcomes in both the RA patients and the general population, alongside its predictive performance compared to other markers. This study comprised 2247 RA patients and a control cohort of 29,177 individuals from the general population. Over a 20-year follow-up period, 738 all-cause deaths and 215 deaths attributable to cardiovascular disease were documented in RA patients. We observed a nonlinear positive correlation between the SII and both all-cause and cardiovascular mortality in RA patients. Of significance, at an SII level of 529.7, the hazard ratio reached 1, signifying a transition from low to high mortality risk. Moreover, subgroup analysis did not reveal any potential interactions. Our study findings indicate a nonlinear positive correlation between the inflammatory biomarker SII and both all-cause and cardiovascular mortality in patients with RA.


Subject(s)
Arthritis, Rheumatoid , Cardiovascular Diseases , Inflammation , Humans , Arthritis, Rheumatoid/mortality , Arthritis, Rheumatoid/immunology , Male , Female , Cardiovascular Diseases/mortality , Cardiovascular Diseases/immunology , Middle Aged , Inflammation/immunology , Retrospective Studies , United States/epidemiology , Adult , Biomarkers , Aged , Prognosis , Proportional Hazards Models , Cause of Death , Nutrition Surveys , ROC Curve , Risk Factors
3.
Int J Mol Sci ; 25(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39000373

ABSTRACT

Atherosclerotic vascular disease disproportionately affects persons living with HIV (PLWH) compared to those without. The reasons for the excess risk include dysregulated immune response and inflammation related to HIV infection itself, comorbid conditions, and co-infections. Here, we review an updated understanding of immune and inflammatory pathways underlying atherosclerosis in PLWH, including effects of viral products, soluble mediators and chemokines, innate and adaptive immune cells, and important co-infections. We also present potential therapeutic targets which may reduce cardiovascular risk in PLWH.


Subject(s)
Atherosclerosis , HIV Infections , Inflammation , Humans , HIV Infections/immunology , HIV Infections/complications , Atherosclerosis/immunology , Inflammation/immunology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/etiology , Animals , Immunity, Innate
4.
FASEB J ; 38(13): e23786, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38979903

ABSTRACT

Regulatory T cells (Tregs) are crucial in regulating T-cell-mediated immune responses. Numerous studies have shown that dysfunction or decreased numbers of Tregs may be involved in inflammatory cardiovascular diseases (CVDs) such as atherosclerosis, hypertension, myocardial infarction, myocarditis, cardiomyopathy, valvular heart diseases, heart failure, and abdominal aortic aneurysm. Tregs can help to ameliorate CVDs by suppressing excessive inflammation through various mechanisms, including inhibition of T cells and B cells, inhibition of macrophage-induced inflammation, inhibition of dendritic cells and foam cell formation, and induction of anti-inflammatory macrophages. Enhancing or restoring the immunosuppressive activity of Tregs may thus serve as a fundamental immunotherapy to treat hypertension and CVDs. However, the precise molecular mechanisms underlying the Tregs-induced protection against hypertension and CVDs remain to be investigated. This review focuses on recent advances in our understanding of Tregs subsets and function in CVDs. In addition, we discuss promising strategies for using Tregs through various pharmacological approaches to treat hypertension and CVDs.


Subject(s)
Cardiovascular Diseases , T-Lymphocytes, Regulatory , Humans , T-Lymphocytes, Regulatory/immunology , Cardiovascular Diseases/immunology , Animals , Inflammation/immunology
5.
Cardiovasc Diabetol ; 23(1): 212, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902748

ABSTRACT

BACKGROUND: This study sought to elucidate the associations of cardiometabolic index (CMI), as a metabolism-related index, with all-cause and cardiovascular mortality among the older population. Utilizing data from the National Health and Nutrition Examination Survey (NHANES), we further explored the potential mediating effect of inflammation within these associations. METHODS: A cohort of 3029 participants aged over 65 years old, spanning six NHANES cycles from 2005 to 2016, was enrolled and assessed. The primary endpoints of the study included all-cause mortality and cardiovascular mortality utilizing data from National Center for Health Statistics (NCHS). Cox regression model and subgroup analysis were conducted to assess the associations of CMI with all-cause and cardiovascular mortality. The mediating effect of inflammation-related indicators including leukocyte, neutrophil, lymphocyte, systemic immune-inflammation index (SII), neutrophil to lymphocyte ratio (NLR) were evaluated to investigate the potential mechanism of the associations between CMI and mortality through mediation package in R 4.2.2. RESULTS: The mean CMI among the enrolled participants was 0.74±0.66, with an average age of 73.28±5.50 years. After an average follow-up period of 89.20 months, there were 1,015 instances of all-cause deaths and 348 cardiovascular deaths documented. In the multivariable-adjusted model, CMI was positively related to all-cause mortality (Hazard Ratio (HR)=1.11, 95% CI=1.01-1.21). Mediation analysis indicated that leukocytes and neutrophils mediated 6.6% and 13.9% of the association of CMI with all-cause mortality. CONCLUSION: Elevated CMI is positively associated with all-cause mortality in the older adults. The association appeared to be partially mediated through inflammatory pathways, indicating that CMI may serve as a valuable indicator for poor prognosis among the older population.


Subject(s)
Cardiometabolic Risk Factors , Cardiovascular Diseases , Cause of Death , Inflammation , Nutrition Surveys , Humans , Male , Aged , Female , Inflammation/blood , Inflammation/mortality , Inflammation/diagnosis , Inflammation/immunology , Cardiovascular Diseases/mortality , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/immunology , Cardiovascular Diseases/blood , Risk Assessment , United States/epidemiology , Aged, 80 and over , Time Factors , Prognosis , Inflammation Mediators/blood , Age Factors , Neutrophils/immunology , Lymphocyte Count , Biomarkers/blood
7.
Acta Physiol (Oxf) ; 240(8): e14193, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38899764

ABSTRACT

The gut-immune axis is a relatively novel phenomenon that provides mechanistic links between the gut microbiome and the immune system. A growing body of evidence supports it is key in how the gut microbiome contributes to several diseases, including hypertension and cardiovascular diseases (CVDs). Evidence over the past decade supports a causal link of the gut microbiome in hypertension and its complications, including myocardial infarction, atherosclerosis, heart failure, and stroke. Perturbations in gut homeostasis such as dysbiosis (i.e., alterations in gut microbial composition) may trigger immune responses that lead to chronic low-grade inflammation and, ultimately, the development and progression of these conditions. This is unsurprising, as the gut harbors one of the largest numbers of immune cells in the body, yet is a phenomenon not entirely understood in the context of cardiometabolic disorders. In this review, we discuss the role of the gut microbiome, the immune system, and inflammation in the context of hypertension and CVD, and consolidate current evidence of this complex interplay, whilst highlighting gaps in the literature. We focus on diet as one of the major modulators of the gut microbiota, and explain key microbial-derived metabolites (e.g., short-chain fatty acids, trimethylamine N-oxide) as potential mediators of the communication between the gut and peripheral organs such as the heart, arteries, kidneys, and the brain via the immune system. Finally, we explore the dual role of both the gut microbiome and the immune system, and how they work together to not only contribute, but also mitigate hypertension and CVD.


Subject(s)
Cardiovascular Diseases , Gastrointestinal Microbiome , Hypertension , Humans , Gastrointestinal Microbiome/physiology , Hypertension/immunology , Hypertension/physiopathology , Hypertension/microbiology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/microbiology , Animals , Dysbiosis/immunology , Inflammation/immunology , Inflammation/metabolism
8.
Front Immunol ; 15: 1344773, 2024.
Article in English | MEDLINE | ID: mdl-38887301

ABSTRACT

Background: Cardiovascular diseases (CVDs) stand as the foremost global cause of mortality, prompting a growing interest in using the potential of immune cells for heart injury treatment. This study aims to assess the causal association between immune cells and CVDs. Methods: A total of 731 immune cells were derived from a previously published genome-wide association study (GWAS), which included approximately 22 million genetic variants among 3,757 individuals of Sardinian ancestry. Genetic associations with atrial fibrillation (AF), heart failure, coronary artery disease, myocardial infarction and stroke were extracted from large-scale GWAS. A two-sample Mendelian randomization (MR) analysis was used to assess the causal association between immune cells and CVDs. Replication MR analysis based on FinnGen dataset and meta-analysis are sequentially conducted to validate causal relationships. Results: Collectively, genetically predicted 4 immune cell traits were associated with AF and 5 immune cell traits were associated with stroke. Increased levels of IgD- CD38dim absolute count were associated with a higher susceptibility to AF, while increased expression of CD14+ CD16+ monocytes, CD62L on CD62L+ myeloid dendritic cells, and CD16 on CD14- CD16+ monocytes were linked to a decreased susceptibility to AF. Additionally, an elevated susceptibility to stroke was linked to an increase in the percentage of CD39+ resting Tregs and heightened CD27 expression on IgD- CD38+ cells. Conversely, a decreased susceptibility to stroke was associated with increased CD40 expression on monocytes, particularly on CD14+ CD16+ and CD14+ CD16- monocytes, with the latter two showing the most compelling evidence. Conclusion: This study identified several immune cell traits that have a causal relationship with CVDs, thus confirming that immune cells play an important role in the pathogenesis of these diseases.


Subject(s)
Cardiovascular Diseases , Genetic Predisposition to Disease , Genome-Wide Association Study , Immunophenotyping , Mendelian Randomization Analysis , Humans , Cardiovascular Diseases/genetics , Cardiovascular Diseases/immunology , Polymorphism, Single Nucleotide
10.
Circ Res ; 134(12): 1663-1680, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843286

ABSTRACT

Over the past 30 years, the field of cardioimmunology has moved from being dismissed as a field that was chasing an epiphenomenon of little biological consequence to a scientific discipline that is providing important new insights into the immunologic basis for hypertension, atherosclerosis, myocarditis, pericarditis, autoimmune heart disease, and heart failure. In this article, we will review the conceptual insights and technical breakthroughs that have allowed the field to move forward, as well as the clinical trials in the cardioimmunology space, to provide a historical context for the articles that will appear in the compendium that is focused on the interface between cardioimmunology, myocardial function, and disease.


Subject(s)
Heart Diseases , Humans , Animals , Heart Diseases/immunology , Heart Diseases/therapy , Allergy and Immunology/trends , Allergy and Immunology/history , Cardiovascular Diseases/immunology , History, 21st Century , History, 20th Century
11.
Arterioscler Thromb Vasc Biol ; 44(7): 1512-1522, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38813699

ABSTRACT

The adaptive immune system plays an important role in the development and progression of atherosclerotic cardiovascular disease. B cells can have both proatherogenic and atheroprotective roles, making treatments aimed at modulating B cells important therapeutic targets. The innate-like B-cell response is generally considered atheroprotective, while the adaptive response is associated with mixed consequences for atherosclerosis. Additionally, interactions of B cells with components of the adaptive and innate immune system, including T cells and complement, also represent key points for therapeutic regulation. In this review, we discuss therapeutic approaches based on B-cell depletion, modulation of B-cell survival, manipulation of both the antibody-dependent and antibody-independent B-cell response, and emerging immunization techniques.


Subject(s)
Adaptive Immunity , B-Lymphocytes , Cardiovascular Diseases , Humans , B-Lymphocytes/immunology , Animals , Cardiovascular Diseases/immunology , Cardiovascular Diseases/therapy , Immunity, Innate , Atherosclerosis/immunology , Atherosclerosis/therapy , Cell Survival
12.
Cytokine Growth Factor Rev ; 77: 91-103, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735805

ABSTRACT

Interleukin-17 (IL-17), a pivotal cytokine in immune regulation, has attracted significant attention in recent years due to its roles in various physiological and pathological processes. This review explores IL-17 in immunological context, emphasizing its structure, production, and signaling pathways. Specifically, we explore its involvement in inflammatory diseases and autoimmune diseases, with a notable focus on its emerging implications in cardiovascular system. Through an array of research insights, IL-17 displays multifaceted functions yet awaiting comprehensive discovery. Highlighting therapeutic avenues, we scrutinize the efficacy and clinical application of four marketed IL-17 mAbs along other targeted therapies, emphasizing their potential in immune-mediated disease management. Additionally, we discussed the novel IL-17D-CD93 axis, elucidating recent breakthroughs in their biological function and clinical implications, inviting prospects for transformative advancements in immunology and beyond.


Subject(s)
Autoimmune Diseases , Cardiovascular Diseases , Interleukin-17 , Humans , Interleukin-17/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Cardiovascular Diseases/immunology , Cardiovascular Diseases/therapy , Signal Transduction , Inflammation/immunology , Cardiovascular System/immunology , Antibodies, Monoclonal/therapeutic use
13.
Immunol Rev ; 324(1): 95-103, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38747455

ABSTRACT

Fat is stored in distinct depots with unique features in both mice and humans and B cells reside in all adipose depots. We have shown that B cells modulate cardiometabolic disease through activities in two of these key adipose depots: visceral adipose tissue (VAT) and perivascular adipose tissue (PVAT). VAT refers to the adipose tissue surrounding organs, within the abdomen and thorax, and is comprised predominantly of white adipocytes. This depot has been implicated in mediating obesity-related dysmetabolism. PVAT refers to adipose tissue surrounding major arteries. It had long been thought to exist to provide protection and insulation for the vessel, yet recent work demonstrates an important role for PVAT in harboring immune cells, promoting their function and regulating the biology of the underlying vessel. The role of B-2 cells and adaptive immunity in adipose tissue biology has been nicely reviewed elsewhere. Given that, the predominance of B-1 cells in adipose tissue at homeostasis, and the emerging role of B-1 cells in a variety of disease states, we will focus this review on how B-1 cells function in VAT and PVAT depots to promote homeostasis and limit inflammation linked to cardiometabolic disease and factors that regulate this function.


Subject(s)
Adipose Tissue , Immunity, Innate , Inflammation , Humans , Animals , Inflammation/immunology , Inflammation/metabolism , Adipose Tissue/metabolism , Adipose Tissue/immunology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Obesity/immunology , Obesity/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Metabolic Diseases/immunology , Metabolic Diseases/metabolism , Metabolic Diseases/etiology , Immunomodulation
14.
Mol Immunol ; 171: 36-46, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763105

ABSTRACT

Damage to the heart can start the repair process and cause cardiac remodeling. B cells play an important role in this process. B cells are recruited to the injured place and activate cardiac remodeling through secreting antibodies and cytokines. Different types of B cells showed specific functions in the heart. Among all types of B cells, heart-associated B cells play a vital role in the heart by secreting TGFß1. B cells participate in the activation of fibroblasts and promote cardiac fibrosis. Four subtypes of B cells in the heart revealed the relationship between the B cells' heterogeneity and cardiac remodeling. Many cardiovascular diseases like atherosclerosis, heart failure (HF), hypertension, myocardial infarction (MI), and dilated cardiomyopathy (DCM) are related to B cells. The primary mechanisms of these B cell-related activities will be discussed in this review, which may also suggest potential novel therapeutic targets.


Subject(s)
B-Lymphocytes , Cardiovascular Diseases , Humans , Cardiovascular Diseases/immunology , Animals , B-Lymphocytes/immunology , Fibrosis/immunology
15.
Pharmacol Res ; 204: 107215, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744399

ABSTRACT

The ubiquitinproteasome system (UPS) is the main mechanism responsible for the intracellular degradation of misfolded or damaged proteins. Under inflammatory conditions, the immunoproteasome, an isoform of the proteasome, can be induced, enhancing the antigen-presenting function of the UPS. Furthermore, the immunoproteasome also serves nonimmune functions, such as maintaining protein homeostasis and regulating signalling pathways, and is involved in the pathophysiological processes of various cardiovascular diseases (CVDs). This review aims to provide a comprehensive summary of the current research on the involvement of the immunoproteasome in cardiovascular diseases, with the ultimate goal of identifying novel strategies for the treatment of these conditions.


Subject(s)
Cardiovascular Diseases , Proteasome Endopeptidase Complex , Humans , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/immunology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Animals , Ubiquitin/metabolism , Ubiquitin/immunology , Signal Transduction
16.
Nature ; 629(8010): 174-183, 2024 May.
Article in English | MEDLINE | ID: mdl-38693412

ABSTRACT

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/ ).


Subject(s)
Endurance Training , Multiomics , Physical Conditioning, Animal , Physical Endurance , Animals , Female , Humans , Male , Rats , Acetylation , Blood/immunology , Blood/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Databases, Factual , Epigenome , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Internet , Lipidomics , Metabolome , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Organ Specificity/genetics , Organ Specificity/immunology , Organ Specificity/physiology , Phosphorylation , Physical Conditioning, Animal/physiology , Physical Endurance/genetics , Physical Endurance/physiology , Proteome/metabolism , Proteomics , Time Factors , Transcriptome/genetics , Ubiquitination , Wounds and Injuries/genetics , Wounds and Injuries/immunology , Wounds and Injuries/metabolism
17.
Clin Immunol ; 264: 110237, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38723855

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) shares several clinical and immunological features with Kawasaki Disease (KD) and pediatric hyperinflammation, but the immuno-phenotypic overlap among these clinical mimics is still incompletely understood. Here we analyzed serum samples from treatment-naïve patients with MIS-C (n = 31) and KD (n = 11), pediatric hyperinflammation (n = 13) and healthy controls (HC, n = 10) by proximity extension assay (PEA) to profile 184 blood biomarkers. Collectively, immunophenotypic overlap between MIS-C and hyperinflammation exceeds overlap with KD. Overexpression of IL-17A in MIS-C and KD could best separate these conditions from hyperinflammatory conditions, while those were hallmarked by overabundance of adenosin deaminase and IL-18. Depletion in serum TNF-related subfamily member 9 (TNFRSF9) and apoptosis inducing ligand (TRAIL) linked with cardiovascular manifestations and myocarditis in MIS-C. Altogether, our analysis highlights important differences in molecular marker signatures also across different MIS-C and KD cohorts and suggests several previously unidentified molecular associations in context of cardiovascular inflammation.


Subject(s)
Biomarkers , Mucocutaneous Lymph Node Syndrome , Proteomics , Systemic Inflammatory Response Syndrome , Humans , Biomarkers/blood , Mucocutaneous Lymph Node Syndrome/blood , Mucocutaneous Lymph Node Syndrome/immunology , Male , Female , Proteomics/methods , Child , Child, Preschool , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/immunology , Inflammation/blood , Infant , Interleukin-17/blood , TNF-Related Apoptosis-Inducing Ligand/blood , Interleukin-18/blood , Adenosine Deaminase/blood , Cardiovascular Diseases/blood , Cardiovascular Diseases/immunology
18.
Front Immunol ; 15: 1402817, 2024.
Article in English | MEDLINE | ID: mdl-38803502

ABSTRACT

Sterile inflammation, characterized by a persistent chronic inflammatory state, significantly contributes to the progression of various diseases such as autoimmune, metabolic, neurodegenerative, and cardiovascular disorders. Recent evidence has increasingly highlighted the intricate connection between inflammatory responses and cardiovascular diseases, underscoring the pivotal role of the Stimulator of Interferon Genes (STING). STING is crucial for the secretion of type I interferon (IFN) and proinflammatory cytokines in response to cytosolic nucleic acids, playing a vital role in the innate immune system. Specifically, research has underscored the STING pathway involvement in unregulated inflammations, where its aberrant activation leads to a surge in inflammatory events, enhanced IFN I responses, and cell death. The primary pathway triggering STING activation is the cyclic GMP-AMP synthase (cGAS) pathway. This review delves into recent findings on STING and the cGAS-STING pathways, focusing on their regulatory mechanisms and impact on cardiovascular diseases. It also discusses the latest advancements in identifying antagonists targeting cGAS and STING, and concludes by assessing the potential of cGAS or STING inhibitors as treatments for cardiovascular diseases.


Subject(s)
Cardiovascular Diseases , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Humans , Nucleotidyltransferases/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/immunology , Membrane Proteins/metabolism , Animals , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism
19.
Circ Res ; 134(11): 1546-1565, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781300

ABSTRACT

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.


Subject(s)
Cardiovascular Diseases , HIV Infections , Humans , HIV Infections/immunology , HIV Infections/epidemiology , HIV Infections/complications , Cardiovascular Diseases/immunology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/diagnostic imaging , Animals
20.
Signal Transduct Target Ther ; 9(1): 130, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38816371

ABSTRACT

The immune response holds a pivotal role in cardiovascular disease development. As multifunctional cells of the innate immune system, macrophages play an essential role in initial inflammatory response that occurs following cardiovascular injury, thereby inducing subsequent damage while also facilitating recovery. Meanwhile, the diverse phenotypes and phenotypic alterations of macrophages strongly associate with distinct types and severity of cardiovascular diseases, including coronary heart disease, valvular disease, myocarditis, cardiomyopathy, heart failure, atherosclerosis and aneurysm, which underscores the importance of investigating macrophage regulatory mechanisms within the context of specific diseases. Besides, recent strides in single-cell sequencing technologies have revealed macrophage heterogeneity, cell-cell interactions, and downstream mechanisms of therapeutic targets at a higher resolution, which brings new perspectives into macrophage-mediated mechanisms and potential therapeutic targets in cardiovascular diseases. Remarkably, myocardial fibrosis, a prevalent characteristic in most cardiac diseases, remains a formidable clinical challenge, necessitating a profound investigation into the impact of macrophages on myocardial fibrosis within the context of cardiac diseases. In this review, we systematically summarize the diverse phenotypic and functional plasticity of macrophages in regulatory mechanisms of cardiovascular diseases and unprecedented insights introduced by single-cell sequencing technologies, with a focus on different causes and characteristics of diseases, especially the relationship between inflammation and fibrosis in cardiac diseases (myocardial infarction, pressure overload, myocarditis, dilated cardiomyopathy, diabetic cardiomyopathy and cardiac aging) and the relationship between inflammation and vascular injury in vascular diseases (atherosclerosis and aneurysm). Finally, we also highlight the preclinical/clinical macrophage targeting strategies and translational implications.


Subject(s)
Cardiovascular Diseases , Macrophages , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Cardiovascular Diseases/therapy , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Fibrosis/genetics , Inflammation/genetics , Inflammation/pathology , Inflammation/immunology , Animals
SELECTION OF CITATIONS
SEARCH DETAIL