Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 535
1.
Circ Res ; 134(12): 1663-1680, 2024 Jun 07.
Article En | MEDLINE | ID: mdl-38843286

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.


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
3.
Front Immunol ; 15: 1365174, 2024.
Article En | MEDLINE | ID: mdl-38774873

Introduction: Sepsis remains a major source of morbidity and mortality in neonates, and characterization of immune regulation in the neonatal septic response remains limited. HVEM is a checkpoint regulator which can both stimulate or inhibit immune responses and demonstrates altered expression after sepsis. We hypothesized that signaling via HVEM would be essential for the neonatal response to sepsis, and that therefore blockade of this pathway would improve survival to septic challenge. Methods: To explore this, neonatal mice were treated with cecal slurry (CS), CS with Anti-HVEM antibody (CS-Ab) or CS with isotype (CS-IT) and followed for 7-day survival. Mice from all treatment groups had thymus, lung, kidney and peritoneal fluid harvested, weighed, and stained for histologic evaluation, and changes in cardiac function were assessed with echocardiography. Results: Mortality was significantly higher for CS-Ab mice (72.2%) than for CS-IT mice (22.2%). CS resulted in dysregulated alveolar remodeling, but CS-Ab lungs demonstrated significantly less dysfunctional alveolar remodeling than CS alone (MCL 121.0 CS vs. 87.6 CS-Ab), as well as increased renal tubular vacuolization. No morphologic differences in alveolar septation or thymic karyorrhexis were found between CS-Ab and CS-IT. CS-Ab pups exhibited a marked decrease in heart rate (390.3 Sh vs. 342.1 CS-Ab), stroke volume (13.08 CS-IT vs. 8.83 CS-Ab) and ultimately cardiac output (4.90 Sh vs. 3.02 CS-Ab) as well as a significant increase in ejection fraction (73.74 Sh vs. 83.75 CS-Ab) and cardiac strain (40.74 Sh vs. 51.16 CS-Ab) as compared to CS-IT or Sham animals. Discussion: While receptor ligation of aspects of HVEM signaling, via antibody blockade, appears to mitigate aspects of lung injury and thymic involution, stimulatory signaling via HVEM still seems to be necessary for vascular and hemodynamic resilience and overall neonatal mouse survival in response to this experimental polymicrobial septic insult. This dissonance in the activity of anti-HVEM neutralizing antibody in neonatal animals speaks to the differences in how septic cardiac dysfunction should be considered and approached in the neonatal population.


Animals, Newborn , Neonatal Sepsis , Signal Transduction , Animals , Mice , Neonatal Sepsis/immunology , Neonatal Sepsis/mortality , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Receptors, Tumor Necrosis Factor, Member 14/immunology , Disease Models, Animal , Female , Heart Diseases/etiology , Heart Diseases/immunology , Lung/immunology , Lung/pathology , Sepsis/immunology , Sepsis/metabolism
5.
Clin Neurol Neurosurg ; 241: 108285, 2024 06.
Article En | MEDLINE | ID: mdl-38636361

BACKGROUND: Stroke-induced heart syndrome is a feared complication of ischemic stroke, that is commonly encountered and has a strong association with unfavorable prognosis. More research is needed to explore underlying mechanisms and inform clinical decision making. This study aims to explore the relationship between the early systemic immune-inflammation (SII) index and the cardiac complications after acute ischemic stroke. METHODS: Consecutive patients with acute ischemic stroke were prospectively collected from January 2020 to August 2022 and retrospectively analyzed. We included subjects who presented within 24 hours after symptom onset and were free of detectable infections or cancer on admission. SII index [(neutrophils × platelets/ lymphocytes)/1000] was calculated from laboratory data at admission. RESULTS: A total of 121 patients were included in our study, of which 24 (19.8 %) developed cardiac complications within 14 days following acute ischemic stroke. The SII level was found higher in patients with stroke-heart syndrome (p<.001), which was an independent predictor of stroke-heart syndrome (adjusted odds ratio 5.089, p=.002). CONCLUSION: New-onset cardiovascular complications diagnosed following a stroke are very common and are associated with early SII index.


Inflammation , Ischemic Stroke , Humans , Male , Female , Ischemic Stroke/immunology , Ischemic Stroke/complications , Retrospective Studies , Aged , Middle Aged , Inflammation/immunology , Heart Diseases/etiology , Heart Diseases/immunology , Heart Diseases/complications , Aged, 80 and over , Brain Ischemia/immunology , Brain Ischemia/complications , Brain Ischemia/etiology
6.
Arterioscler Thromb Vasc Biol ; 44(6): 1181-1190, 2024 Jun.
Article En | MEDLINE | ID: mdl-38634279

Cardiovascular diseases remain the largest cause of death worldwide with recent evidence increasingly attributing the development and progression of these diseases to an exacerbated inflammatory response. As a result, significant research is now focused on modifying the immune environment to prevent the disease progression. This in turn has highlighted the lymphatic system in the pathophysiology of cardiovascular diseases owing, in part, to its established function in immune cell surveillance and trafficking. In this review, we highlight the role of the cardiac lymphatic system and its potential as an immunomodulatory therapeutic target in selected cardiovascular diseases.


Lymphatic Vessels , Humans , Animals , Lymphatic Vessels/physiopathology , Lymphatic Vessels/immunology , Lymphatic Vessels/metabolism , Heart Diseases/physiopathology , Heart Diseases/immunology , Heart Diseases/pathology , Heart Diseases/metabolism , Heart Diseases/therapy , Signal Transduction , Lymphangiogenesis , Lymphatic System/physiopathology , Lymphatic System/immunology
7.
Int J Mol Sci ; 24(23)2023 Nov 21.
Article En | MEDLINE | ID: mdl-38068879

Inflammation and inflammasomes have been proposed as important regulators of the host-microorganism interaction, playing a key role in morbidity and mortality due to the coronavirus disease 2019 (COVID-19) in subjects with chronic conditions and compromised immune system. The inflammasome consists of a multiprotein complex that finely regulates the activation of caspase-1 and the production and secretion of potent pro-inflammatory cytokines such as IL-1ß and IL-18. The pyrin containing NOD (nucleotide-binding oligomerization domain) like receptor (NLRP) is a family of intracellular receptors, sensing patterns associated to pathogens or danger signals and NLRP3 inflammasome is the most deeply analyzed for its involvement in the innate and adaptive immune system as well as its contribution to several autoinflammatory and autoimmune diseases. It is highly expressed in leukocytes and up-regulated in sentinel cells upon inflammatory stimuli. NLRP3 expression has also been reported in B and T lymphocytes, in epithelial cells of oral and genital mucosa, in specific parenchymal cells as cardiomyocytes, and keratinocytes, and chondrocytes. It is well known that a dysregulated activation of the inflammasome is involved in the pathogenesis of different disorders that share the common red line of inflammation in their pathogenetic fingerprint. Here, we review the potential roles of the NLRP3 inflammasome in cardiovascular events, liver damage, pulmonary diseases, and in that wide range of systemic inflammatory syndromes named as a cytokine storm.


Cytokine Release Syndrome , Heart Diseases , Inflammasomes , Liver Diseases , Lung Diseases , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Carrier Proteins/metabolism , Cytokine Release Syndrome/immunology , Inflammasomes/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Lung Diseases/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Heart Diseases/immunology , Liver Diseases/immunology
8.
Eur J Pharmacol ; 913: 174644, 2021 Dec 15.
Article En | MEDLINE | ID: mdl-34801532

The mortality of sepsis-induced cardiac dysfunction (SICD) is very high due to the complex pathophysiological mechanism. Syringaresinol (SYR) is a natural abstract which possesses anti-inflammatory property. The present study aims was to identify the protective impact of SYR on sepsis-induced cardiac dysfunction and investigate the specific mechanisms. We found that SYR improved the cardiac function and alleviated myocardial injury in mice that subjected to cecal ligation and puncture, in addition, SIRT1 expression was significantly elevated after SYR treatment compared to sepsis group both in vivo and in vitro, along with suppression of NLRP3 activation and proinflammatory cytokines release. However, SIRT1 inhibitor EX427 abolished the impact of SYR on LPS-induced pyroptosis in cardiomyocytes. Furthermore, molecular docking analysis predicted that there is high affinity between SYR and estrogen receptor (ER), ER inhibitor ICI182780, the specific ERß inhibitor PHTP and the specific ERαinhibitor AZD9496 were used to examine the role of ER in the protective effect of SYR against SICD, and the results suggested that ER activation was essential for the cardioprotective function of SYR. In conclusion, SYR ameliorates SICD via the ER/SIRT1/NLRP3/GSDMD pathway.


Cardiotonic Agents/pharmacology , Furans/pharmacology , Heart Diseases/drug therapy , Lignans/pharmacology , Receptors, Estrogen/metabolism , Sepsis/complications , Animals , Cardiotonic Agents/therapeutic use , Cinnamates/pharmacology , Disease Models, Animal , Fulvestrant/pharmacology , Furans/therapeutic use , Heart/drug effects , Heart Diseases/immunology , Heart Diseases/pathology , Humans , Indoles/pharmacology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Lignans/therapeutic use , Male , Mice , Molecular Docking Simulation , Myocardium/pathology , Pyroptosis/drug effects , Pyroptosis/immunology , Receptors, Estrogen/antagonists & inhibitors , Sepsis/drug therapy , Sepsis/immunology , Sirtuin 1/metabolism
9.
Melo, Marcelo Dantas Tavares de; Paiva, Marcelo Goulart; Santos, Maria Verônica Câmara; Rochitte, Carlos Eduardo; Moreira, Valéria de Melo; Saleh, Mohamed Hassan; Soares, Brandão, Simone Cristina; Gallafrio, Claudia Cosentino; Goldwasser, Daniel; Gripp, Eliza de Almeida; Piveta, Rafael Bonafim; Silva, Tonnison Oliveira; Santo, Thais Harada Campos Espirito; Ferreira, Waldinai Pereira; Salemi, Vera Maria Cury; Cauduro, Sanderson A; Barberato, Silvio Henrique; Lopes, Heloísa M Christovam; Pena, José Luiz Barros; Rached, Heron Rhydan Saad; Miglioranza, Marcelo Haertel; Pinheiro, Aurélio Carvalho; Vrandecic, Bárbara Athayde Linhares Martins; Cruz, Cecilia Beatriz Bittencourt Viana; Nomura, César Higa; Cerbino, Fernanda Mello Erthal; Costa, Isabela Bispo Santos da Silva; Coelho-Filho, Otavio Rizzi; Carneiro, Adriano Camargo de Castro; Burgos, Ursula Maria Moreira Costa; Fernandes, Juliano Lara; Uellendahl, Marly; Calado, Eveline Barros; Senra, Tiago; Assunção, Bruna Leal; Freire, Claudia Maria Vilas; Martins, Cristiane Nunes; Sawamura, Karen Saori Shiraishi; Brito, Márcio Miranda; Jardim, Maria Fernanda Silva; Bernardes, Renata Junqueira Moll; Diógenes, Tereza Cristina; Vieira, Lucas de Oliveira; Mesquita, Claudio Tinoco; Lopes, Rafael Willain; Neto, Elry Medeiros Vieira Segundo; Rigo, Letícia; Marin, Valeska Leite Siqueira; Santos, Marcelo José; Grossman, Gabriel Blacher; Quagliato, Priscila Cestari; Alcantara, Monica Luiza de; Teodoro, José Aldo Ribeiro; Albricker, Ana Cristina Lopes; Barros, Fanilda Souto; Amaral, Salomon Israel do; Porto, Carmen Lúcia Lascasas; Barros, Marcio Vinícius Lins; Santos, Simone Nascimento dos; Cantisano, Armando Luís; Petisco, Ana Cláudia Gomes Pereira; Barbosa, José Eduardo Martins; Veloso, Orlando Carlos Glória; Spina, Salvador; Pignatelli, Ricardo; Hajjar, Ludhmilla Abrahão; Filho, Roberto Kalil; Lopes, Marcelo Antônio Cartaxo Queiroga; Vieira, Marcelo Luiz Campos; Almeida, André Luiz Cerqueira.
Arq. bras. cardiol ; 117(4): 845-909, Oct. 2021. graf, ilus, tab
Article Pt | CONASS, SES-SP, SESSP-IDPCPROD, SES-SP | ID: biblio-1344557
10.
Viruses ; 13(10)2021 09 24.
Article En | MEDLINE | ID: mdl-34696354

Viruses are an underappreciated cause of heart failure. Indeed, several types of viral infections carry cardiovascular risks. Understanding shared and unique mechanisms by which each virus compromises heart function is critical to inform on therapeutic interventions. This review describes how the key viruses known to lead to cardiac dysfunction operate. Both direct host-damaging mechanisms and indirect actions on the immune systems are discussed. As viral myocarditis is a key pathologic driver of heart failure in infected individuals, this review also highlights the role of cytokine storms and inflammation in virus-induced cardiomyopathy.


Heart Failure/virology , Heart/virology , Myocarditis/virology , Animals , Cardiomyopathies/virology , Cardiomyopathy, Dilated/virology , Cytokine Release Syndrome , Heart Diseases/immunology , Heart Diseases/therapy , Heart Diseases/virology , Heart Failure/immunology , Heart Failure/therapy , Humans , Inflammation , Myocarditis/immunology , Myocarditis/therapy , Virus Diseases/immunology , Virus Diseases/therapy , Virus Diseases/virology
11.
Front Endocrinol (Lausanne) ; 12: 726967, 2021.
Article En | MEDLINE | ID: mdl-34484128

In March 2020, the WHO declared coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a global pandemic. Obesity was soon identified as a risk factor for poor prognosis, with an increased risk of intensive care admissions and mechanical ventilation, but also of adverse cardiovascular events. Obesity is associated with adipose tissue, chronic low-grade inflammation, and immune dysregulation with hypertrophy and hyperplasia of adipocytes and overexpression of pro-inflammatory cytokines. However, to implement appropriate therapeutic strategies, exact mechanisms must be clarified. The role of white visceral adipose tissue, increased in individuals with obesity, seems important, as a viral reservoir for SARS-CoV-2 via angiotensin-converting enzyme 2 (ACE2) receptors. After infection of host cells, the activation of pro-inflammatory cytokines creates a setting conducive to the "cytokine storm" and macrophage activation syndrome associated with progression to acute respiratory distress syndrome. In obesity, systemic viral spread, entry, and prolonged viral shedding in already inflamed adipose tissue may spur immune responses and subsequent amplification of a cytokine cascade, causing worse outcomes. More precisely, visceral adipose tissue, more than subcutaneous fat, could predict intensive care admission; and lower density of epicardial adipose tissue (EAT) could be associated with worse outcome. EAT, an ectopic adipose tissue that surrounds the myocardium, could fuel COVID-19-induced cardiac injury and myocarditis, and extensive pneumopathy, by strong expression of inflammatory mediators that could diffuse paracrinally through the vascular wall. The purpose of this review is to ascertain what mechanisms may be involved in unfavorable prognosis among COVID-19 patients with obesity, especially cardiovascular events, emphasizing the harmful role of excess ectopic adipose tissue, particularly EAT.


COVID-19/metabolism , Cardiomyopathies/metabolism , Intra-Abdominal Fat/metabolism , Obesity/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/complications , COVID-19/immunology , Cardiomyopathies/immunology , Cardiomyopathies/pathology , Heart Diseases/immunology , Heart Diseases/metabolism , Heart Diseases/pathology , Humans , Inflammation , Intra-Abdominal Fat/pathology , Obesity/complications , Obesity/immunology , Obesity/pathology , Pericardium , Prognosis , SARS-CoV-2/metabolism , Serine Endopeptidases/metabolism
12.
Front Immunol ; 12: 737170, 2021.
Article En | MEDLINE | ID: mdl-34512671

Cerebral-cardiac syndrome (CCS) refers to cardiac dysfunction following varying brain injuries. Ischemic stroke is strongly evidenced to induce CCS characterizing as arrhythmia, myocardial damage, and heart failure. CCS is attributed to be the second leading cause of death in the post-stroke stage; however, the responsible mechanisms are obscure. Studies indicated the possible mechanisms including insular cortex injury, autonomic imbalance, catecholamine surge, immune response, and systemic inflammation. Of note, the characteristics of the stroke population reveal a common comorbidity with diabetes. The close and causative correlation of diabetes and stroke directs the involvement of diabetes in CCS. Nevertheless, the role of diabetes and its corresponding molecular mechanisms in CCS have not been clarified. Here we conclude the features of CCS and the potential role of diabetes in CCS. Diabetes drives establish a "primed" inflammatory microenvironment and further induces severe systemic inflammation after stroke. The boosted inflammation is suspected to provoke cardiac pathological changes and hence exacerbate CCS. Importantly, as the key element of inflammation, NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome is indicated to play an important role in diabetes, stroke, and the sequential CCS. Overall, we characterize the corresponding role of diabetes in CCS and speculate a link of NLRP3 inflammasome between them.


Diabetes Mellitus/immunology , Heart Diseases/immunology , Inflammasomes/immunology , Inflammation Mediators/immunology , Ischemic Stroke/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Comorbidity , Diabetes Mellitus/epidemiology , Diabetes Mellitus/metabolism , Diabetes Mellitus/physiopathology , Heart Diseases/epidemiology , Heart Diseases/metabolism , Heart Diseases/physiopathology , Humans , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Ischemic Stroke/epidemiology , Ischemic Stroke/metabolism , Ischemic Stroke/physiopathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction , Syndrome
13.
Cells ; 10(7)2021 07 03.
Article En | MEDLINE | ID: mdl-34359851

Chronic inflammation, the activation of immune cells and their cross-talk with cardiomyocytes in the pathogenesis and progression of heart diseases has long been overlooked. However, with the latest research developments, it is increasingly accepted that a vicious cycle exists where cardiomyocytes release cardiocrine signaling molecules that spiral down to immune cell activation and chronic state of low-level inflammation. For example, cardiocrine molecules released from injured or stressed cardiomyocytes can stimulate macrophages, dendritic cells, neutrophils and even T-cells, which then subsequently increase cardiac inflammation by co-stimulation and positive feedback loops. One of the key proteins involved in stress-mediated cardiomyocyte signal transduction is a small GTPase RhoA. Importantly, the regulation of RhoA activation is critical for effective immune cell response and is being considered as one of the potential therapeutic targets in many immune-cell-mediated inflammatory diseases. In this review we provide an update on the role of RhoA at the juncture of immune cell activation, inflammation and cardiac disease.


Heart Diseases/immunology , Leukocytes/immunology , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Animals , Cardiomegaly/immunology , Cardiomegaly/pathology , Heart Diseases/pathology , Heart Failure/immunology , Heart Failure/pathology , Humans
14.
J Am Heart Assoc ; 10(16): e021428, 2021 08 17.
Article En | MEDLINE | ID: mdl-34365798

Background Cardiac dysfunction is a prominent feature of multisystem inflammatory syndrome in children (MIS-C), yet the etiology is poorly understood. We determined whether dysfunction is global or regional, and whether it is associated with the cytokine milieu, microangiopathy, or severity of shock. Methods and Results We analyzed echocardiographic parameters of myocardial deformation and compared global and segmental left ventricular strain between 43 cases with MIS-C ≤18 years old and 40 controls. Primary outcomes included left ventricular global longitudinal strain, right ventricular free wall strain), and left atrial strain. We evaluated relationships between strain and profiles of 10 proinflammatory cytokines, microangiopathic features (soluble C5b9), and vasoactive-inotropic requirements. Compared with controls, cases with MIS-C had significant impairments in all parameters of systolic and diastolic function. 65% of cases with MIS-C had abnormal left ventricular function (|global longitudinal strain|<17%), although elevations of cytokines were modest. All left ventricular segments were involved, without apical or basal dominance to suggest acute stress cardiomyopathy. Worse global longitudinal strain correlated with higher ratios of interleukin-6 (ρ -0.43) and interleukin-8 (ρ -0.43) to total hypercytokinemia, but not absolute levels of interleukin-6 or interleukin-8, or total hypercytokinemia. Similarly, worse right ventricular free wall strain correlated with higher relative interleukin-8 expression (ρ -0.59). There were no significant associations between function and microangiopathy or vasoactive-inotropic requirements. Conclusions Myocardial function is globally decreased in MIS-C and not explained by acute stress cardiomyopathy. Cardiac dysfunction may be driven by the relative skew of the immune response toward interleukin-6 and interleukin-8 pathways, more so than degree of hyperinflammation, refining the current paradigm of myocardial involvement in MIS-C.


Atrial Function, Left , COVID-19/complications , Cytokine Release Syndrome/etiology , Cytokines/blood , Heart Diseases/etiology , Inflammation Mediators/blood , Systemic Inflammatory Response Syndrome/complications , Ventricular Function, Left , Ventricular Function, Right , Adolescent , Age Factors , Biomarkers/blood , COVID-19/diagnosis , COVID-19/immunology , Child , Cross-Sectional Studies , Cytokine Release Syndrome/diagnosis , Cytokine Release Syndrome/immunology , Echocardiography , Female , Heart Diseases/diagnostic imaging , Heart Diseases/immunology , Heart Diseases/physiopathology , Humans , Male , Prognosis , Retrospective Studies , Risk Assessment , Risk Factors , Systemic Inflammatory Response Syndrome/diagnosis , Systemic Inflammatory Response Syndrome/immunology
15.
Cardiovasc Pathol ; 54: 107370, 2021.
Article En | MEDLINE | ID: mdl-34273507

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is commonly associated with myocardial injury and heart failure. The pathophysiology behind this phenomenon remains unclear, with many diverse and multifaceted hypotheses. To contribute to this understanding, we describe the underlying cardiac findings in fifty patients who died with coronavirus disease 2019 (COVID-19). METHODS: Included were autopsies performed on patients with a positive SARS-CoV-2 reverse-transcriptase-polymerase-chain reaction test from the index hospitalization. In the case of out-of-hospital death, patients were included if post-mortem testing was positive. Complete autopsies were performed according to a COVID-19 safety protocol, and all patients underwent both macroscopic and microscopic examination. If available, laboratory findings and echocardiograms were reported. RESULTS: The median age of the decedents was 63.5 years. The most common comorbidities included hypertension (90.0%), diabetes (56.0%) and obesity (50.0%). Lymphocytic inflammatory infiltrates in the heart were present in eight (16.0%) patients, with focal myocarditis present in two (4.0%) patients. Acute myocardial ischemia was observed in eight (16.0%) patients. The most common findings were myocardial fibrosis (80.0%), hypertrophy (72.0%), and microthrombi (66.0%). The most common causes of death were COVID-19 pneumonia in 18 (36.0%), COVID-19 pneumonia with bacterial superinfection in 12 (24.0%), and COVID-19 pneumonia with pulmonary embolism in 10 (20.0%) patients. CONCLUSIONS: Cardiovascular comorbidities were prevalent, and pathologic changes associated with hypertensive and atherosclerotic cardiovascular disease were the most common findings. Despite markedly elevated inflammatory markers and cardiac enzymes, few patients exhibited inflammatory infiltrates or necrosis within cardiac myocytes. A unifying pathophysiologic mechanism behind myocardial injury in COVID-19 remains elusive, and additional autopsy studies are needed.


COVID-19/pathology , Heart Diseases/pathology , Myocardium/pathology , SARS-CoV-2/pathogenicity , Adult , Aged , Aged, 80 and over , Atherosclerosis/mortality , Atherosclerosis/pathology , Autopsy , COVID-19/immunology , COVID-19/mortality , COVID-19/virology , Comorbidity , Female , Heart Diseases/immunology , Heart Diseases/mortality , Heart Diseases/virology , Host-Pathogen Interactions , Humans , Hypertension/mortality , Hypertension/pathology , Inflammation Mediators/analysis , Male , Middle Aged , Myocardium/immunology , Necrosis , SARS-CoV-2/immunology , Up-Regulation
17.
Pharmacol Res ; 168: 105581, 2021 06.
Article En | MEDLINE | ID: mdl-33781873

In-depth characterization of heart-brain communication in critically ill patients with severe acute respiratory failure is attracting significant interest in the COronaVIrus Disease 19 (COVID-19) pandemic era during intensive care unit (ICU) stay and after ICU or hospital discharge. Emerging research has provided new insights into pathogenic role of the deregulation of the heart-brain axis (HBA), a bidirectional flow of information, in leading to severe multiorgan disease syndrome (MODS) in patients with confirmed infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Noteworthy, HBA dysfunction may worsen the outcome of the COVID-19 patients. In this review, we discuss the critical role HBA plays in both promoting and limiting MODS in COVID-19. We also highlight the role of HBA as new target for novel therapeutic strategies in COVID-19 in order to open new translational frontiers of care. This is a translational perspective from the Italian Society of Cardiovascular Researches.


Brain Diseases/therapy , Brain/drug effects , COVID-19/therapy , Heart Diseases/therapy , Heart/drug effects , Adrenal Cortex Hormones/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Antiviral Agents/administration & dosage , Brain/immunology , Brain/metabolism , Brain Diseases/immunology , Brain Diseases/metabolism , COVID-19/immunology , COVID-19/metabolism , Critical Care/methods , Critical Illness/therapy , Dietary Supplements , Functional Food , Heart Diseases/immunology , Heart Diseases/metabolism , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Microvessels/drug effects , Microvessels/immunology , Microvessels/metabolism , Multiple Organ Failure/immunology , Multiple Organ Failure/metabolism , Multiple Organ Failure/therapy , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , SARS-CoV-2/metabolism
18.
Front Immunol ; 12: 584538, 2021.
Article En | MEDLINE | ID: mdl-33679735

The cardiovascular and immune systems undergo profound and intertwined alterations with aging. Recent studies have reported that an accumulation of memory and terminally differentiated T cells in elderly subjects can fuel myocardial aging and boost the progression of heart diseases. Nevertheless, it remains unclear whether the immunological senescence profile is sufficient to cause age-related cardiac deterioration or merely acts as an amplifier of previous tissue-intrinsic damage. Herein, we sought to decompose the causality in this cardio-immune crosstalk by studying young mice harboring a senescent-like expanded CD4+ T cell compartment. Thus, immunodeficient NSG-DR1 mice expressing HLA-DRB1*01:01 were transplanted with human CD4+ T cells purified from matching donors that rapidly engrafted and expanded in the recipients without causing xenograft reactions. In the donor subjects, the CD4+ T cell compartment was primarily composed of naïve cells defined as CCR7+CD45RO-. However, when transplanted into young lymphocyte-deficient mice, CD4+ T cells underwent homeostatic expansion, upregulated expression of PD-1 receptor and strongly shifted towards effector/memory (CCR7- CD45RO+) and terminally-differentiated phenotypes (CCR7-CD45RO-), as typically seen in elderly. Differentiated CD4+ T cells also infiltrated the myocardium of recipient mice at comparable levels to what is observed during physiological aging. In addition, young mice harboring an expanded CD4+ T cell compartment showed increased numbers of infiltrating monocytes, macrophages and dendritic cells in the heart. Bulk mRNA sequencing analyses further confirmed that expanding T-cells promote myocardial inflammaging, marked by a distinct age-related transcriptomic signature. Altogether, these data indicate that exaggerated CD4+ T-cell expansion and differentiation, a hallmark of the aging immune system, is sufficient to promote myocardial alterations compatible with inflammaging in juvenile healthy mice.


Aging/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Heart Diseases/immunology , Immunologic Memory/immunology , Myocardium/immunology , Aging/genetics , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cells, Cultured , Gene Expression/immunology , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/immunology , HLA-DRB1 Chains/metabolism , Heart Diseases/genetics , Heart Diseases/metabolism , Humans , Immunologic Memory/genetics , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , RNA-Seq/methods , Transplantation, Heterologous
19.
Immunobiology ; 226(2): 152071, 2021 03.
Article En | MEDLINE | ID: mdl-33588306

Dendritic cell-associated C-type lectin-1 (Dectin-1), also known as ß-glucan receptor is an emerging pattern recognition receptor (PRR) which belongs to the family of C-type lectin receptor (CLR). This CLR identifies ligands independently of Ca2+ and is majorly involved in coupling of innate with adaptive immunity. Formerly, Dectin-1 was best known for its role in anti-fungal defense only. However, recent explorations suggested its wider role in defense against variety of infectious diseases caused by pathogens including bacteria, parasites and viruses. In fact, Dectin-1 signaling axis has been suggested to be targeted as an effective therapeutic strategy for cancers. Dectin-1 has also been elucidated ascetically in the heart, respiratory, intestinal, neurological and developmental disorders. Being a defensive PRR, Dectin-1 results in optimal immune responses in collaboration with other PRRs, but the overall evaluation reinforces the hypothesis of disease development on dis-regulation of Dectin-1 activity. This underscores the impact of Dectin-1 polymorphisms in modulating protein expression and generation of non-optimal immune responses through defective collaborations, further underlining their therapeutic potential. To add on, Dectin-1 influence autoimmunity and severe inflammation accredited to recognition of self T cells and apoptotic cells through unknown ligands. Few reports have also testified its redundant role in infections, which makes it a complicated molecule to be fully resolved. Thus, Dectin-1 is a hub that runs a complex collaborative network, whose interactive wire connections to different PRRs are still pending to be revealed. Alternatively, so far focus of almost all the researchers was the two major cell surface isoforms of Dectin-1, despite the fact that its soluble functional intracellular isoform (Dectin-1E) has already been dissected but is indefinable. Therefore, this review intensely recommends the need of future research to resolve the un-resolved and treasure the comprehensive role of Dectin-1 in different clinical outcomes, before determining its therapeutic prospective.


Lectins, C-Type/immunology , Receptors, Pattern Recognition/immunology , Animals , Autoimmune Diseases/immunology , Autophagy , Heart Diseases/immunology , Humans , Infections/immunology , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Neoplasms/immunology , Nervous System Diseases/immunology , Receptors, Pattern Recognition/chemistry , Receptors, Pattern Recognition/genetics , Respiratory Tract Diseases/immunology
20.
Am J Med Sci ; 361(6): 718-724, 2021 06.
Article En | MEDLINE | ID: mdl-33582156

BACKGROUND: Inflammation can facilitate development of coronavirus disease 2019 (COVID-19) and cardiac injury is associated with worse clinical outcomes. However, data are relatively scarce on the association between hyper-inflammatory response and cardiac injury among COVID-19 patients. METHODS: The study was designed based on severe and critically ill patients with COVID-19. Information on clinical characteristics and laboratory examinations was collected from the electronic medical records and analyzed. RESULTS: There were 32.4% (n = 107) of patients with cardiac injury. The median age was 67 years, and 48.8% (n = 161) of patients were men. Hypertension was the most common in 161 (48.8%) patients, followed by diabetes (16.7%, n = 55) and coronary heart disease (13.3%, n = 44). Compared to cases without cardiac injury, those with cardiac injury were older, had higher proportions of coronary heart disease, and leukocyte counts, significantly elevated concentrations of N-terminal pro-B-Type natriuretic peptide, high-sensitivity C-reactive protein (hs-CRP), tumor necrosis factor (TNF)-α, interleukin-2 receptor (IL-2R), IL-6, and IL-8, but lower lymphocyte counts. A significant positive correlation was observed between high-sensitivity troponin I and inflammatory cytokines. Logistic regression analysis showed that hs-CRP, TNF-α and IL-6 were independent risk factors for cardiac injury. CONCLUSIONS: Cardiac injury was associated with elevated levels of inflammatory cytokines among severe and critically ill patients with COVID-19, suggesting that hyper-inflammatory response may involve in cardiac injury.


COVID-19 , Heart Diseases , SARS-CoV-2 , Troponin I/blood , Aged , C-Reactive Protein/analysis , COVID-19/immunology , COVID-19/physiopathology , COVID-19/therapy , Cardiometabolic Risk Factors , China/epidemiology , Critical Illness/epidemiology , Critical Illness/therapy , Diabetes Mellitus/epidemiology , Female , Heart Diseases/diagnosis , Heart Diseases/immunology , Heart Diseases/virology , Humans , Hypertension/epidemiology , Interleukin-6/blood , Male , Risk Assessment , SARS-CoV-2/isolation & purification , SARS-CoV-2/pathogenicity , Severity of Illness Index , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/complications , Systemic Inflammatory Response Syndrome/virology , Tumor Necrosis Factor-alpha/blood
...