Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Circulation ; 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38506045

RESUMO

BACKGROUND: Viral infections can cause acute respiratory distress syndrome (ARDS), systemic inflammation, and secondary cardiovascular complications. Lung macrophage subsets change during ARDS, but the role of heart macrophages in cardiac injury during viral ARDS remains unknown. Here we investigate how immune signals typical for viral ARDS affect cardiac macrophage subsets, cardiovascular health, and systemic inflammation. METHODS: We assessed cardiac macrophage subsets using immunofluorescence histology of autopsy specimens from 21 patients with COVID-19 with SARS-CoV-2-associated ARDS and 33 patients who died from other causes. In mice, we compared cardiac immune cell dynamics after SARS-CoV-2 infection with ARDS induced by intratracheal instillation of Toll-like receptor ligands and an ACE2 (angiotensin-converting enzyme 2) inhibitor. RESULTS: In humans, SARS-CoV-2 increased total cardiac macrophage counts and led to a higher proportion of CCR2+ (C-C chemokine receptor type 2 positive) macrophages. In mice, SARS-CoV-2 and virus-free lung injury triggered profound remodeling of cardiac resident macrophages, recapitulating the clinical expansion of CCR2+ macrophages. Treating mice exposed to virus-like ARDS with a tumor necrosis factor α-neutralizing antibody reduced cardiac monocytes and inflammatory MHCIIlo CCR2+ macrophages while also preserving cardiac function. Virus-like ARDS elevated mortality in mice with pre-existing heart failure. CONCLUSIONS: Our data suggest that viral ARDS promotes cardiac inflammation by expanding the CCR2+ macrophage subset, and the associated cardiac phenotypes in mice can be elicited by activating the host immune system even without viral presence in the heart.

2.
Cell Rep ; 42(2): 112046, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36708514

RESUMO

The diversity of mononuclear phagocyte (MNP) subpopulations across tissues is one of the key physiological characteristics of the immune system. Here, we focus on understanding the metabolic variability of MNPs through metabolic network analysis applied to three large-scale transcriptional datasets: we introduce (1) an ImmGen MNP open-source dataset of 337 samples across 26 tissues; (2) a myeloid subset of ImmGen Phase I dataset (202 MNP samples); and (3) a myeloid mouse single-cell RNA sequencing (scRNA-seq) dataset (51,364 cells) assembled based on Tabula Muris Senis. To analyze such large-scale datasets, we develop a network-based computational approach, genes and metabolites (GAM) clustering, for unbiased identification of the key metabolic subnetworks based on transcriptional profiles. We define 9 metabolic subnetworks that encapsulate the metabolic differences within MNP from 38 different tissues. Obtained modules reveal that cholesterol synthesis appears particularly active within the migratory dendritic cells, while glutathione synthesis is essential for cysteinyl leukotriene production by peritoneal and lung macrophages.


Assuntos
Fagócitos , Análise de Célula Única , Animais , Camundongos
3.
Nat Cardiovasc Res ; 1(3): 263-280, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35959412

RESUMO

Heart failure represents a major cause of morbidity and mortality worldwide. Single-cell transcriptomics have revolutionized our understanding of cell composition and associated gene expression. Through integrated analysis of single-cell and single-nucleus RNA-sequencing data generated from 27 healthy donors and 18 individuals with dilated cardiomyopathy, here we define the cell composition of the healthy and failing human heart. We identify cell-specific transcriptional signatures associated with age and heart failure and reveal the emergence of disease-associated cell states. Notably, cardiomyocytes converge toward common disease-associated cell states, whereas fibroblasts and myeloid cells undergo dramatic diversification. Endothelial cells and pericytes display global transcriptional shifts without changes in cell complexity. Collectively, our findings provide a comprehensive analysis of the cellular and transcriptomic landscape of human heart failure, identify cell type-specific transcriptional programs and disease-associated cell states and establish a valuable resource for the investigation of human heart failure.

4.
Circulation ; 145(10): 765-782, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35113652

RESUMO

BACKGROUND: Recent studies have established that CCR2 (C-C chemokine receptor type 2) marks proinflammatory subsets of monocytes, macrophages, and dendritic cells that contribute to adverse left ventricle (LV) remodeling and heart failure progression. Elucidation of the effector mechanisms that mediate adverse effects of CCR2+ monocytes, macrophages, and dendritic cells will yield important insights into therapeutic strategies to suppress myocardial inflammation. METHODS: We used mouse models of reperfused myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation to investigate CCL17 (C-C chemokine ligand 17). We used Ccl17 knockout mice, flow cytometry, RNA sequencing, biochemical assays, cell trafficking studies, and in vivo cell depletion to identify the cell types that generate CCL17, define signaling pathways that controlled its expression, delineate the functional importance of CCL17 in adverse LV remodeling and heart failure progression, and determine the mechanistic basis by which CCL17 exerts its effects. RESULTS: We demonstrated that CCL17 is expressed in CCR2+ macrophages and cluster of differentiation 11b+ conventional dendritic cells after myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation. We clarified the transcriptional signature of CCL17+ macrophages and dendritic cells and identified granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling as a key regulator of CCL17 expression through cooperative activation of STAT5 (signal transducer and activator of transcription 5) and canonical NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells) signaling. Ccl17 deletion resulted in reduced LV remodeling, decreased myocardial fibrosis and cardiomyocyte hypertrophy, and improved LV systolic function after myocardial infarction and angiotensin II and phenylephrine infusion. We observed increased abundance of regulatory T cells (Tregs) in the myocardium of injured Ccl17 knockout mice. CCL17 inhibited Treg recruitment through biased activation of CCR4. CCL17 activated Gq signaling and CCL22 (C-C chemokine ligand 22) activated both Gq and ARRB (ß-arrestin) signaling downstream of CCR4. CCL17 competitively inhibited CCL22 stimulated ARRB signaling and Treg migration. We provide evidence that Tregs mediated the protective effects of Ccl17 deletion on myocardial inflammation and adverse LV remodeling. CONCLUSIONS: These findings identify CCL17 as a proinflammatory mediator of CCR2+ macrophages and dendritic cells and suggest that inhibition of CCL17 may serve as an effective strategy to promote Treg recruitment and suppress myocardial inflammation.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Angiotensina II/farmacologia , Animais , Quimiocina CCL17/metabolismo , Quimiocina CCL17/farmacologia , Toxina Diftérica/metabolismo , Toxina Diftérica/farmacologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Humanos , Inflamação/metabolismo , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenilefrina/metabolismo , Fenilefrina/farmacologia , Linfócitos T Reguladores/metabolismo , Remodelação Ventricular
5.
Immunity ; 54(9): 2072-2088.e7, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34320366

RESUMO

Cardiac macrophages represent a heterogeneous cell population with distinct origins, dynamics, and functions. Recent studies have revealed that C-C Chemokine Receptor 2 positive (CCR2+) macrophages derived from infiltrating monocytes regulate myocardial inflammation and heart failure pathogenesis. Comparatively little is known about the functions of tissue resident (CCR2-) macrophages. Herein, we identified an essential role for CCR2- macrophages in the chronically failing heart. Depletion of CCR2- macrophages in mice with dilated cardiomyopathy accelerated mortality and impaired ventricular remodeling and coronary angiogenesis, adaptive changes necessary to maintain cardiac output in the setting of reduced cardiac contractility. Mechanistically, CCR2- macrophages interacted with neighboring cardiomyocytes via focal adhesion complexes and were activated in response to mechanical stretch through a transient receptor potential vanilloid 4 (TRPV4)-dependent pathway that controlled growth factor expression. These findings establish a role for tissue-resident macrophages in adaptive cardiac remodeling and implicate mechanical sensing in cardiac macrophage activation.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Ativação de Macrófagos/fisiologia , Macrófagos/metabolismo , Remodelação Ventricular/fisiologia , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Miocárdio/metabolismo , Troponina T/genética
6.
Immunity ; 54(6): 1110-1122, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34107270

RESUMO

The interindividual heterogeneity of the immune system likely determines the personal risk for acquiring infections and developing diseases with inflammatory components. In addition to genetic factors, the immune system's heterogeneity is driven by diverging exposures of leukocytes and their progenitors to infections, vaccinations, and health behavior, including lifestyle-related stimuli such as diet, physical inactivity, and psychosocial stress. We review how such experiences alter immune cell responses to concurrent and subsequent challenges, leading to either improved host resilience or disease susceptibility due to a muted or overzealous immune system, with a primary focus on the contribution of innate immune cells. We explore the involvement of diverse mechanisms, including trained immunity, and their relevance for infections and cardiovascular disease, as these prevalent conditions are heavily influenced by immune cell abundance and phenotypic adaptions. Understanding the mechanistic bases of immune modulations by prior or co-exposures may lead to new therapies targeting dysfunctional inflammation.


Assuntos
Doenças Cardiovasculares/imunologia , Suscetibilidade a Doenças/imunologia , Sistema Imunitário/imunologia , Infecções/imunologia , Animais , Humanos , Imunidade Inata/imunologia , Inflamação/imunologia , Estilo de Vida
7.
J Nucl Med ; 62(1): 111-114, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32444372

RESUMO

Proinflammatory macrophages are important mediators of inflammation after myocardial infarction and of allograft injury after heart transplantation. The aim of this study was to image the recruitment of proinflammatory chemokine receptor 2-positive (CCR2+) cells in multiple heart injury models. Methods:64Cu-DOTA-extracellular loop 1 inverso (ECL1i) PET was used to image CCR2+ monocytes and macrophages in a heart transplantation mouse model. Flow cytometry was performed to characterize CCR2+ cells. Autoradiography on a human heart specimen was conducted to confirm binding specificity. 64Cu- and 68Ga-DOTA-ECL1i were compared in an ischemia-reperfusion injury mouse model. Results:64Cu-DOTA-ECL1i showed sensitive and specific detection of CCR2+ cells in all tested mouse models, with efficacy comparable to that of 68Ga-DOTA-ECL1i. Flow cytometry demonstrated specific expression of CCR2 on monocytes and macrophages. The tracer binds to human CCR2. Conclusion: This work establishes the utility of 64Cu-DOTA-ECL1i to image CCR2+ monocytes and macrophages in mouse models and provides the requisite preclinical information to translate the targeted clinical-grade CCR2 imaging probe for clinical investigation of heart diseases.


Assuntos
Traumatismos Cardíacos/diagnóstico por imagem , Traumatismos Cardíacos/metabolismo , Monócitos/metabolismo , Femprocumona/metabolismo , Tomografia por Emissão de Pósitrons , Receptores CCR2/metabolismo , Animais , Marcação por Isótopo , Camundongos , Camundongos Endogâmicos C57BL
8.
JCI Insight ; 4(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31672943

RESUMO

Lysosomes are at the epicenter of cellular processes critical for inflammasome activation in macrophages. Inflammasome activation and IL-1ß secretion are implicated in myocardial infarction (MI) and resultant heart failure; however, little is known about how macrophage lysosomes regulate these processes. In mice subjected to cardiac ischemia/reperfusion (IR) injury and humans with ischemic cardiomyopathy, we observed evidence of lysosomal impairment in macrophages. Inducible macrophage-specific overexpression of transcription factor EB (TFEB), a master regulator of lysosome biogenesis (Mϕ-TFEB), attenuated postinfarction remodeling, decreased abundance of proinflammatory macrophages, and reduced levels of myocardial IL-1ß compared with controls. Surprisingly, neither inflammasome suppression nor Mϕ-TFEB-mediated attenuation of postinfarction myocardial dysfunction required intact ATG5-dependent macroautophagy (hereafter termed "autophagy"). RNA-seq of flow-sorted macrophages postinfarction revealed that Mϕ-TFEB upregulated key targets involved in lysosomal lipid metabolism. Specifically, inhibition of the TFEB target, lysosomal acid lipase, in vivo abrogated the beneficial effect of Mϕ-TFEB on postinfarction ventricular function. Thus, TFEB reprograms macrophage lysosomal lipid metabolism to attenuate remodeling after IR, suggesting an alternative paradigm whereby lysosome function affects inflammation.


Assuntos
Proteína 5 Relacionada à Autofagia/fisiologia , Autofagia/fisiologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Macrófagos/metabolismo , Infarto do Miocárdio/fisiopatologia , Disfunção Ventricular , Animais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
JCI Insight ; 52019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31162139

RESUMO

Whereas prior studies have demonstrated an important immunomodulatory role for the neuronal cholinergic system in the heart, the role of the non-neuronal cholinergic system is not well understood. To address the immunomodulatory role of the non-neuronal cholinergic system in the heart we used a previously validated diphtheria toxin (DT)-induced cardiomyocyte ablation model (Rosa26-DTMlc2v-Cre mice). DT-injected Rosa26-DTMlc2v-Cre mice were treated with diluent or Pyridostigmine Bromide (PYR), a reversible cholinesterase inhibitor. PYR treatment resulted in increased survival and decreased numbers of MHC-IIlowCCR2+ macrophages in DT-injected Rosa26-DTMlc2v-Cre mice compared to diluent treated Rosa26-DTMlc2v-Cre mice. Importantly, the expression of CCL2/7 mRNA and protein was reduced in the hearts of PYR-treated mice. Backcrossing Rosa26-DTMlc2v-Cre mice with a transgenic mouse line (Chat-ChR2) that constitutively overexpresses the vesicular acetylcholine transporter (VAChT) resulted in decreased expression of Ccl2/7 mRNA and decreased numbers of CD68+ cells in DT-injured Rosa26-DTMlc2v-Cre/Chat-ChR2 mouse hearts, consistent with the pharmacologic studies with PYR. In vitro studies with cultures of LPS-stimulated peritoneal macrophages revealed a concentration-dependent reduction in CCL2 secretion following stimulation with ACh, nicotine and muscarine. Viewed together, these findings reveal a previously unappreciated immunomodulatory role for the non-neuronal cholinergic system in regulating homeostatic responses in the heart following tissue injury.


Assuntos
Colinérgicos/imunologia , Colinérgicos/metabolismo , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/microbiologia , Miócitos Cardíacos/metabolismo , Neurônios/metabolismo , Animais , Quimiocina CCL2/metabolismo , Quimiocina CCL7/metabolismo , Quimiocinas/metabolismo , Toxina Diftérica/efeitos adversos , Modelos Animais de Doenças , Feminino , Homeostase , Inflamação/imunologia , Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , RNA Mensageiro/metabolismo , Proteínas Vesiculares de Transporte de Acetilcolina
10.
Circ Res ; 124(6): 881-890, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30661445

RESUMO

RATIONALE: Paradigm shifting studies have revealed that the heart contains functionally diverse populations of macrophages derived from distinct embryonic and adult hematopoietic progenitors. Under steady-state conditions, the heart is largely populated by CCR2- (C-C chemokine receptor type 2) macrophages of embryonic descent. After tissue injury, a dramatic shift in macrophage composition occurs whereby CCR2+ monocytes are recruited to the heart and differentiate into inflammatory CCR2+ macrophages that contribute to heart failure progression. Currently, there are no techniques to noninvasively detect CCR2+ monocyte recruitment into the heart and thus identify patients who may be candidates for immunomodulatory therapy. OBJECTIVE: To develop a noninvasive molecular imaging strategy with high sensitivity and specificity to visualize inflammatory monocyte and macrophage accumulation in the heart. METHODS AND RESULTS: We synthesized and tested the performance of a positron emission tomography radiotracer (68Ga-DOTA [1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid]-ECL1i [extracellular loop 1 inverso]) that allosterically binds to CCR2. In naive mice, the radiotracer was quickly cleared from the blood and displayed minimal retention in major organs. In contrast, biodistribution and positron emission tomography demonstrated strong myocardial tracer uptake in 2 models of cardiac injury (diphtheria toxin induced cardiomyocyte ablation and reperfused myocardial infarction). 68Ga-DOTA-ECL1i signal localized to sites of tissue injury and was independent of blood pool activity as assessed by quantitative positron emission tomography and ex vivo autoradiography. 68Ga-DOTA-ECL1i uptake was associated with CCR2+ monocyte and CCR2+ macrophage infiltration into the heart and was abrogated in CCR2-/- mice, demonstrating target specificity. Autoradiography demonstrated that 68Ga-DOTA-ECL1i specifically binds human heart failure specimens and with signal intensity associated with CCR2+ macrophage abundance. CONCLUSIONS: These findings demonstrate the sensitivity and specificity of 68Ga-DOTA-ECL1i in the mouse heart and highlight the translational potential of this agent to noninvasively visualize CCR2+ monocyte recruitment and inflammatory macrophage accumulation in patients.


Assuntos
Coração/diagnóstico por imagem , Macrófagos/fisiologia , Monócitos/fisiologia , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/patologia , Animais , Movimento Celular , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Imagem Molecular , Tomografia por Emissão de Pósitrons , Receptores CCR2/análise
11.
J Vis Exp ; (154)2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31904009

RESUMO

Macrophages represent the most heterogeneous and abundant immune cell populations in the heart and are central in driving inflammation and reparative responses after cardiac injury. How various subsets of macrophages orchestrate the immune responses after cardiac injury is an active area of research. Presented here is a simple protocol that our lab performs routinely, for the extraction of macrophages from mouse and human myocardium specimens obtained from healthy and diseased individuals. Briefly, this protocol involves enzymatic digestion of cardiac tissue to generate a single cell suspension, followed by antibody staining, and flow cytometry. This technique is suitable for functional assays performed on sorted cells as well as bulk and single cell RNA sequencing. A major advantage of this protocol is its simplicity, minimal day to day variation and wide applicability allowing investigation of macrophage heterogeneity across various mouse models and human disease entities.


Assuntos
Separação Celular/métodos , Macrófagos/citologia , Miocárdio/citologia , Animais , Citometria de Fluxo , Coração Auxiliar , Humanos , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Células Estromais/citologia
12.
Circ Res ; 124(2): 263-278, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30582448

RESUMO

RATIONALE: Recent advancements have brought to light the origins, complexity, and functions of tissue-resident macrophages. However, in the context of tissue injury or disease, large numbers of monocytes infiltrate the heart and are thought to contribute to adverse remodeling and heart failure pathogenesis. Little is understood about the diversity of monocytes and monocyte-derived macrophages recruited to the heart after myocardial injury, including the mechanisms that regulate monocyte recruitment and fate specification. OBJECTIVE: We sought to test the hypothesis that distinct subsets of tissue-resident CCR2- (C-C chemokine receptor 2) and CCR2+ macrophages orchestrate monocyte recruitment and fate specification after myocardial injury. METHODS AND RESULTS: We reveal that in numerous mouse models of cardiomyocyte cell death (permanent myocardial infarction, reperfused myocardial infarction, and diphtheria toxin cardiomyocyte ablation), there is a shift in macrophage ontogeny whereby tissue-resident macrophages are predominately replaced by infiltrating monocytes and monocyte-derived macrophages. Using syngeneic cardiac transplantation to model ischemia-reperfusion injury and distinguish tissue-resident from recruited cell populations in combination with intravital 2-photon microscopy, we demonstrate that monocyte recruitment is differentially orchestrated by distinct subsets of tissue-resident cardiac macrophages. Tissue-resident CCR2+ macrophages promote monocyte recruitment through an MYD88 (myeloid differentiation primary response 88)-dependent mechanism that results in release of MCPs (monocyte chemoattractant proteins) and monocyte mobilization. In contrast, tissue-resident CCR2- macrophages inhibit monocyte recruitment. Using CD (cluster of differentiation) 169-DTR (diphtheria toxin receptor) and CCR2-DTR mice, we further show that selective depletion of either tissue-resident CCR2- or CCR2+ macrophages before myocardial infarction results in divergent effects on left ventricular function, myocardial remodeling, and monocyte recruitment. Finally, using single-cell RNA sequencing, we show that tissue-resident cardiac macrophages differentially instruct monocyte fate specification. CONCLUSIONS: Collectively, these observations establish the mechanistic basis by which monocytes are initially recruited to the injured heart and provide new insights into the heterogeneity of monocyte-derived macrophages.


Assuntos
Linhagem da Célula , Quimiotaxia de Leucócito , Macrófagos/metabolismo , Monócitos/metabolismo , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Receptores CCR2/metabolismo , Animais , Morte Celular , Toxina Diftérica/farmacologia , Modelos Animais de Doenças , Transplante de Coração , Ativação de Macrófagos , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Fator 88 de Diferenciação Mieloide/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/patologia , Receptores CCR2/genética , Transdução de Sinais , Função Ventricular Esquerda , Remodelação Ventricular
13.
Neurol India ; 66(4): 1020-1027, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30038086

RESUMO

BACKGROUND: Restless legs syndrome (RLS) is misdiagnosed due to a variety of clinical presentations and lack of a diagnostic biomarker. Sociocultural differences in patients' reporting of symptoms further contribute to this under diagnosis. We developed an expanded diagnostic tool for RLS, incorporating all International RLS Study Group (IRLSSG) diagnostic criteria with a number of additional questions mainly focusing on specific sociocultural influences in RLS symptom reporting among Indians. The purpose of this study was to examine the change in the diagnostic yield of RLS, if any, through administration of this expanded questionnaire. MATERIALS AND METHODS: The AIIMS RLS questionnaire for Indian patients (ARQIP) was developed in English language, and then translated into Hindi. All consecutive patients attending Neurology and sleep disorders clinic with complaints of leg discomfort were recruited in the study. Two examiners evaluated all patients with complaints of leg discomfort seen by a senior Sleep Medicine expert, one using only IRLSSG diagnostic criteria and the other using the ARQIP. Patients were categorized as RLS or "no-RLS" by the expert, and this was considered as the "standard" for analysis. RESULTS: A total of 155 participants (78 males, 50.3%) with a mean age of 44.1 ± 14.5 years were enrolled. A total of 105 patients were diagnosed as having RLS (group 1) and the rest as having "non-RLS" (group 2). The ARQIP was found to have a much higher sensitivity (100% vs 73%), specificity (44% vs 32.7%), negative predictive value (100% vs 36.4%), and positive predictive value (79% vs 70%) compared to the standard questionnaire. The diagnostic yield of this tool was 26.7% (Confidence interval = 100-73.3). CONCLUSIONS: The ARQIP for RLS diagnosis, validated in this study, has been observed to have a high sensitivity and a negative predictive value with a high diagnostic accuracy.


Assuntos
Síndrome das Pernas Inquietas/diagnóstico , Inquéritos e Questionários , Traduções , Adulto , Povo Asiático , Feminino , Humanos , Índia , Idioma , Masculino , Pessoa de Meia-Idade , Sensibilidade e Especificidade
14.
Nat Med ; 24(8): 1234-1245, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29892064

RESUMO

Paradigm-shifting studies in the mouse have identified tissue macrophage heterogeneity as a critical determinant of immune responses. In contrast, surprisingly little is known regarding macrophage heterogeneity in humans. Macrophages within the mouse heart are partitioned into CCR2- and CCR2+ subsets with divergent origins, repopulation mechanisms, and functions. Here, we demonstrate that the human myocardium also contains distinct subsets of CCR2- and CCR2+ macrophages. Analysis of sex-mismatched heart transplant recipients revealed that CCR2- macrophages are a tissue-resident population exclusively replenished through local proliferation, whereas CCR2+ macrophages are maintained through monocyte recruitment and proliferation. Moreover, CCR2- and CCR2+ macrophages have distinct functional properties, analogous to reparative CCR2- and inflammatory CCR2+ macrophages in the mouse heart. Clinically, CCR2+ macrophage abundance is associated with left ventricular remodeling and systolic function in heart failure patients. Collectively, these observations provide initial evidence for the functional importance of macrophage heterogeneity in the human heart.


Assuntos
Macrófagos/citologia , Macrófagos/metabolismo , Miocárdio/citologia , Adulto , Insuficiência Cardíaca/patologia , Humanos , Inflamação/patologia , Receptores CCR2/metabolismo , Disfunção Ventricular Esquerda/patologia , Suporte de Carga
15.
JCI Insight ; 3(11)2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29875326

RESUMO

Despite the long-standing recognition that the immune response to acute myocardial injury contributes to adverse left ventricular (LV) remodeling, it has not been possible to effectively target this clinically. Using 2 different in vivo models of acute myocardial injury, we show that pirfenidone confers beneficial effects in the murine heart through an unexpected mechanism that depends on cardiac B lymphocytes. Naive hearts contained a large population of CD19+CD11b-CD23-CD21-IgD+IgMlo lymphocytes, and 2 smaller populations of CD19+CD11b+ B1a and B1b cells. In response to tissue injury, there was an increase in neutrophils, monocytes, macrophages, as well as an increase in CD19+ CD11b- B lymphocytes. Treatment with pirfenidone had no effect on the number of neutrophils, monocytes, or macrophages, but decreased CD19+CD11b- lymphocytes. B cell depletion abrogated the beneficial effects of pirfenidone. In vitro studies demonstrated that stimulation with lipopolysaccharide and extracts from necrotic cells activated CD19+ lymphocytes through a TIRAP-dependent pathway. Treatment with pirfenidone attenuated this activation of B cells. These findings reveal a previously unappreciated complexity of myocardial B lymphocytes within the inflammatory infiltrate triggered by cardiac injury and suggest that pirfenidone exerts beneficial effects in the heart through a unique mechanism that involves modulation of cardiac B lymphocytes.


Assuntos
Subpopulações de Linfócitos B/imunologia , Ventrículos do Coração/efeitos dos fármacos , Infarto do Miocárdio/imunologia , Piridonas/administração & dosagem , Remodelação Ventricular/efeitos dos fármacos , Animais , Subpopulações de Linfócitos B/efeitos dos fármacos , Toxina Diftérica/administração & dosagem , Toxina Diftérica/imunologia , Modelos Animais de Doenças , Feminino , Ventrículos do Coração/imunologia , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Humanos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Ativação Linfocitária/efeitos dos fármacos , Depleção Linfocítica/métodos , Camundongos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/citologia , Miocárdio/imunologia , Miocárdio/patologia , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Remodelação Ventricular/imunologia
16.
JCI Insight ; 2(14)2017 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-28724792

RESUMO

Pediatric dilated cardiomyopathy (DCM) is the most common indication for heart transplantation in children. Despite similar genetic etiologies, medications routinely used in adult heart failure patients do not improve outcomes in the pediatric population. The mechanistic basis for these observations is unknown. We hypothesized that pediatric and adult DCM comprise distinct pathological entities, in that children do not undergo adverse remodeling, the target of adult heart failure therapies. To test this hypothesis, we examined LV specimens obtained from pediatric and adult donor controls and DCM patients. Consistent with the established pathophysiology of adult heart failure, adults with DCM displayed marked cardiomyocyte hypertrophy and myocardial fibrosis compared with donor controls. In contrast, pediatric DCM specimens demonstrated minimal cardiomyocyte hypertrophy and myocardial fibrosis compared with both age-matched controls and adults with DCM. Strikingly, RNA sequencing uncovered divergent gene expression profiles in pediatric and adult patients, including enrichment of transcripts associated with adverse remodeling and innate immune activation in adult DCM specimens. Collectively, these findings reveal that pediatric and adult DCM represent distinct pathological entities, provide a mechanistic basis to explain why children fail to respond to adult heart failure therapies, and suggest the need to develop new approaches for pediatric DCM.

17.
Ann Indian Acad Neurol ; 19(4): 499-504, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27994361

RESUMO

BACKGROUND: Since a majority of population in India does not drive automobiles, one item on the Epworth Sleepiness Scale (ESS) requires modification and validation. In addition, data collected by us indicated that a majority of rural and urban Indians regularly spend time in prayer/spiritual activity. The main purpose of this study was to develop a cross-cultural adaptation of the ESS for a North Indian population, in Hindi language (ESS-I). The study also provides evidence of reliability and validity of the modified version. METHODOLOGY: The subjects included were normal volunteers aged 18-75 years (Group 1) (n = 70), compared with patients with complaints of excessive daytime sleepiness, who had undergone polysomnography (Group 2) (n = 22) and patients who had undergone multiple sleep latency test (Group 3) (n = 10). The study was carried out in four phases: Translation and retranslation of the original scale with modification of item 8 (mainly addition of option of question on "while offering prayers or in spiritual activity"); reliability (test-retest) (n = 30); internal consistency (using Cronbach's alpha index) (n = 102); and sensitivity to change (n = 8). RESULTS: Group 1 showed spiritual activity as a significantly more commonly practiced activity than driving. The Cronbach's alpha for the modified version was 0.892 (excellent), and this was not improved by removing the modified item. The alpha value for Group 1 versus Groups 2 and 3 was 0.667 and 0.892, respectively. The scale was reliable over time (test-retest), and it was sensitive to sleepiness change in patients with obstructive sleep apnea during treatment. CONCLUSION: The ESS-I, is comparable to the original scale. It is reliable, valid, and change-sensitive. It is proposed that the modified version can be very useful for detecting sleepiness among Indian population, especially those who do not drive their own vehicles.

18.
Mol Biosyst ; 10(4): 806-12, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24457312

RESUMO

Adipocytes from white adipose tissue secrete cytokines and other bioactive proteins which are collectively termed adipokines. Adiposity has been linked with increased breast cancer risk as adipokines secreted by adipocytes significantly affect epithelial cells from which breast cancer arises. Measurement of extracellular adipokine concentrations that would be involved in signaling through mammary tissue is therefore of importance. In this work, microdialysis sampling was used to collect adipokines from the interstitial space of the mammary fat pad of female rats under isoflurane anesthesia. The adipokines CCL2 (MCP-1), leptin and IL-6 were quantified from dialysate samples and compared to total tissue concentrations surrounding the implanted probes. After three hours of microdialysis sampling at 1 µL min(-1), the respective median values for these adipokines in dialysate samples were approximately 175 pg mL(-1) (CCL2), 150 pg mL(-1) (IL-6) and 750 pg mL(-1) (leptin). Adipokine protein levels from dialysates were an order of magnitude lower than levels obtained directly from mammary tissue. However, the adipokine concentrations between excised tissue surrounding the microdialysis sampling probes and control tissue without implants did not differ. This work demonstrates the utility of microdialysis sampling to quantify mammary gland adipokine levels, with relevance to understanding mammary physiology.


Assuntos
Tecido Adiposo/metabolismo , Quimiocina CCL2/metabolismo , Interleucina-6/metabolismo , Leptina/metabolismo , Glândulas Mamárias Animais/metabolismo , Adipócitos/metabolismo , Adipocinas/metabolismo , Tecido Adiposo/citologia , Animais , Feminino , Microdiálise , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...