Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78.952
Filtrar
1.
Front Immunol ; 15: 1327372, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38736889

RESUMEN

Introduction: Growing evidence from animal models indicates that the myocardium hosts a population of B cells that play a role in the development of cardiomyopathy. However, there is minimal data on human myocardial B cells in the context of cardiomyopathy. Methods: We integrated single-cell and single-nuclei datasets from 45 healthy human hearts, 70 hearts with dilated cardiomyopathy (DCM), and 8 hearts with arrhythmogenic right ventricular cardiomyopathy (ARVC). Interactions between B cells and other cell types were investigated using the CellChat Package. Differential gene expression analysis comparing B cells across conditions was performed using DESeq2. Pathway analysis was performed using Ingenuity, KEGG, and GO pathways analysis. Results: We identified 1,100 B cells, including naive B cells and plasma cells. Cells showed an extensive network of interactions within the healthy myocardium that included outgoing signaling to macrophages, T cells, endothelial cells, and pericytes, and incoming signaling from endothelial cells, pericytes, and fibroblasts. This niche relied on ECM-receptor, contact, and paracrine interactions; and changed significantly in the context of cardiomyopathy, displaying disease-specific features. Differential gene expression analysis showed that in the context of DCM both naive and plasma B cells upregulated several pathways related to immune activation, including upregulation of oxidative phosphorylation, upregulation of leukocyte extravasation, and, in naive B cells, antigen presentation. Discussion: The human myocardium contains naive B cells and plasma cells, integrated into a diverse and dynamic niche that has distinctive features in healthy, DCM, and ARVC. Naive myocardial-associated B cells likely contribute to the pathogenesis of human DCM.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica , Linfocitos B , Cardiomiopatía Dilatada , Miocardio , Humanos , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/genética , Displasia Ventricular Derecha Arritmogénica/genética , Displasia Ventricular Derecha Arritmogénica/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Miocardio/metabolismo , Miocardio/inmunología , Miocardio/patología , Masculino , Femenino , Comunicación Celular/inmunología , Perfilación de la Expresión Génica , Persona de Mediana Edad , Adulto , Transcriptoma , Regulación de la Expresión Génica
2.
Cardiovasc Toxicol ; 24(6): 527-538, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38720122

RESUMEN

Adolescents commonly co-abuse many drugs including anabolic androgenic steroids either they are athletes or non-athletes. Stanozolol is the major anabolic used in recent years and was reported grouped with cannabis. The current study aimed at evaluating the biochemical and histopathological changes related to the hypertrophic effects of stanozolol and/or cannabis whether in condition of exercise practice or sedentary conditions. Adult male Wistar albino rats received either stanozolol (5 mg/kg, s.c), cannabis (10 mg/kg, i.p.), and a combination of both once daily for two months. Swimming exercise protocol was applied as a training model. Relative heart weight, oxidative stress biomarkers, cardiac tissue fibrotic markers were evaluated. Left ventricular morphometric analysis and collagen quantification was done. The combined treatment exhibited serious detrimental effects on the heart tissues. It increased heart tissue fibrotic markers (Masson's trichrome stain (p < 0.001), cardiac COL3 (p < 0.0001), and VEGF-A (p < 0.05)), lowered heart glutathione levels (p < 0.05) and dramatically elevated oxidative stress (increased malondialdehyde (p < 0.0001) and 8-OHDG (p < 0.0001)). Training was not ameliorating for the observed effects. Misuse of cannabis and stanozolol resulted in more hypertrophic consequences of the heart than either drug alone, which were at least largely assigned to oxidative stress, heart tissue fibrotic indicators, histological alterations, and morphometric changes.


Asunto(s)
Anabolizantes , Cardiomegalia Inducida por el Ejercicio , Fibrosis , Estrés Oxidativo , Ratas Wistar , Estanozolol , Animales , Estanozolol/toxicidad , Masculino , Estrés Oxidativo/efectos de los fármacos , Anabolizantes/toxicidad , Cardiomegalia Inducida por el Ejercicio/efectos de los fármacos , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Izquierda/inducido químicamente , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/prevención & control , Remodelación Ventricular/efectos de los fármacos , Miocardio/patología , Miocardio/metabolismo , Doping en los Deportes , Biomarcadores/metabolismo , Natación , Condicionamiento Físico Animal/fisiología , Ratas , Modelos Animales de Enfermedad
3.
Aging (Albany NY) ; 16(9): 7553-7577, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38742935

RESUMEN

After menopause, the incidence of cardiovascular disease rapidly rises in women. The disappearing protection provided by sex steroids is a consequence of the development of many risk factors. Preclinical studies are necessary to understand better the effects of ovarian hormones loss cardiac aging. To mimic menopause in mice and study its consequences, we delayed ovariectomy at 12 months and followed animals for 12 months. Using RNA sequencing, we investigated changes in the myocardial exome with aging. In addition, with four-core genotypes (FCG) transgenic mice, we studied sex chromosome effects on cardiac aging. Heart weight increased from 3 to 24 months (males + 35%, females + 29%). In males, 75% of this increase had occurred at 12 months; in females, only 30%. Gonadectomy of mice at 12 months blocked cardiac hypertrophy in both sexes during the second year of life. The dosage of the X chromosomes did not influence cardiac growth in young and older mice. We performed an RNA sequencing study in young and old mice. We identified new highly expressed genes modulated during aging (Bdh, Myot, Cpxm2, and Slc38a1). The myocardial exome in older animals displayed few differences related to the animal's sex or the presence or absence of sex steroids for a year. We show that the morphological evolution of the heart depends on the biological sex via gonadal sex hormone actions. The myocardial exome of old male and female mice is relatively similar. Our study emphasizes the need to consider sex steroid effects in studying cardiac aging.


Asunto(s)
Envejecimiento , Hormonas Esteroides Gonadales , Cromosomas Sexuales , Animales , Femenino , Masculino , Envejecimiento/genética , Ratones , Hormonas Esteroides Gonadales/metabolismo , Cromosomas Sexuales/genética , Ratones Transgénicos , Ovariectomía , Corazón , Miocardio/metabolismo , Miocardio/patología , Factores Sexuales , Cardiomegalia/genética
4.
Med Sci Monit ; 30: e945188, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38775003

RESUMEN

This publication has been retracted by the Editor due to the identification of non-original figure images and manuscript content that raise concerns regarding the credibility and originality of the study and the manuscript. Reference: Ying-Jun Zhang, He Huang, Yu Liu, Bin Kong, Guangji Wang. MD-1 Deficiency Accelerates Myocardial Inflammation and Apoptosis in Doxorubicin-Induced Cardiotoxicity by Activating the TLR4/MAPKs/Nuclear Factor kappa B (NF-kappaB) Signaling Pathway. Med Sci Monit, 2019; 25: 7898-7907. DOI: 10.12659/MSM.919861.


Asunto(s)
Apoptosis , Cardiotoxicidad , Doxorrubicina , FN-kappa B , Transducción de Señal , Receptor Toll-Like 4 , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/deficiencia , FN-kappa B/metabolismo , Doxorrubicina/efectos adversos , Doxorrubicina/farmacología , Apoptosis/efectos de los fármacos , Animales , Cardiotoxicidad/metabolismo , Cardiotoxicidad/etiología , Transducción de Señal/efectos de los fármacos , Inflamación/metabolismo , Inflamación/patología , Miocardio/patología , Miocardio/metabolismo , Ratones , Antígeno 96 de los Linfocitos/metabolismo , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo
5.
Sci Adv ; 10(19): eadl3549, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38718121

RESUMEN

Metabolic reprogramming is critical in the onset of pressure overload-induced cardiac remodeling. Our study reveals that proline dehydrogenase (PRODH), the key enzyme in proline metabolism, reprograms cardiomyocyte metabolism to protect against cardiac remodeling. We induced cardiac remodeling using transverse aortic constriction (TAC) in both cardiac-specific PRODH knockout and overexpression mice. Our results indicate that PRODH expression is suppressed after TAC. Cardiac-specific PRODH knockout mice exhibited worsened cardiac dysfunction, while mice with PRODH overexpression demonstrated a protective effect. In addition, we simulated cardiomyocyte hypertrophy in vitro using neonatal rat ventricular myocytes treated with phenylephrine. Through RNA sequencing, metabolomics, and metabolic flux analysis, we elucidated that PRODH overexpression in cardiomyocytes redirects proline catabolism to replenish tricarboxylic acid cycle intermediates, enhance energy production, and restore glutathione redox balance. Our findings suggest PRODH as a modulator of cardiac bioenergetics and redox homeostasis during cardiac remodeling induced by pressure overload. This highlights the potential of PRODH as a therapeutic target for cardiac remodeling.


Asunto(s)
Ratones Noqueados , Miocitos Cardíacos , Prolina , Remodelación Ventricular , Animales , Prolina/metabolismo , Miocitos Cardíacos/metabolismo , Ratones , Ratas , Prolina Oxidasa/metabolismo , Prolina Oxidasa/genética , Metabolismo Energético , Miocardio/metabolismo , Miocardio/patología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/etiología , Modelos Animales de Enfermedad , Oxidación-Reducción , Masculino , Reprogramación Metabólica
7.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 666-674, 2024 Apr 20.
Artículo en Chino | MEDLINE | ID: mdl-38708499

RESUMEN

OBJECTIVE: To investigate the changes of mitochondrial respiratory function during myocardial fibrosis in mice with myocardial infarction (MI) and its correlation with the increase of glycolytic flux. METHODS: Forty C57BL/6N mice were randomized into two equal groups to receive sham operation or ligation of the left anterior descending coronary artery to induce acute MI. At 28 days after the operation, 5 mice from each group were euthanized and left ventricular tissue samples were collected for transcriptomic sequencing. FPKM method was used to calculate gene expression levels to identify the differentially expressed genes (DEGs) in MI mice, which were analyzed using GO and KEGG databases to determine the pathways affecting the disease process. Heat maps were drawn to show the differential expressions of the pathways and the related genes in the enrichment analysis. In primary cultures of neonatal mouse cardiac fibroblasts (CFs), the changes in mitochondrial respiration and glycolysis levels in response to treatment with the pro-fibrotic agonist TGF-ß1 were analyzed using Seahorse experiment. RESULTS: The mouse models of MI showed significantly increased diastolic and systolic left ventricular diameter (P < 0.05) and decreased left ventricular ejection fraction (P < 0.0001). A total of 124 up-regulated and 106 down-regulated DEGs were identified in the myocardial tissues of MI mice, and GO and KEGG enrichment analysis showed that these DEGs were significantly enriched in fatty acid metabolism, organelles and other metabolic pathways and in the mitochondria. Heat maps revealed fatty acid beta oxidation, mitochondrial dysfunction and increased glycolysis levels in MI mice. In the primary culture of CFs, treatment with TGF-ß1 significantly reduced the basal and maximum respiratory levels and increased the basal and maximum glycolysis levels (P < 0.0001). CONCLUSION: During myocardial fibrosis, energy metabolism remodeling occurs in the CFs, manifested by lowered mitochondrial function and increased energy generation through glycolysis.


Asunto(s)
Metabolismo Energético , Fibrosis , Ratones Endogámicos C57BL , Infarto del Miocardio , Animales , Ratones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/genética , Miocardio/metabolismo , Miocardio/patología , Modelos Animales de Enfermedad , Mitocondrias/metabolismo , Glucólisis , Perfilación de la Expresión Génica , Transcriptoma , Fibroblastos/metabolismo , Masculino , Factor de Crecimiento Transformador beta1/metabolismo
8.
Sci Rep ; 14(1): 10365, 2024 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710778

RESUMEN

Cardiac fibroblasts (CFs) are essential for preserving myocardial integrity and function. They can detect variations in cardiac tissue stiffness using various cellular mechanosensors, including the Ca2+ permeable mechanosensitive channel Piezo1. Nevertheless, how CFs adapt the mechanosensitive response to stiffness changes remains unclear. In this work we adopted a multimodal approach, combining the local mechanical stimulation (from 10 pN to 350 nN) with variations of culture substrate stiffness. We found that primary rat CFs cultured on stiff (GPa) substrates showed a broad Piezo1 distribution in the cell with particular accumulation at the mitochondria membrane. CFs displayed a force-dependent behavior in both calcium uptake and channel activation probability, showing a threshold at 300 nN, which involves both cytosolic and mitochondrial Ca2+ mobilization. This trend decreases as the myofibroblast phenotype within the cell population increases, following a possible Piezo1 accumulation at focal adhesion sites. In contrast, the inhibition of fibroblasts to myofibroblasts transition with soft substrates (kPa) considerably reduces both mechanically- and chemically-induced Piezo1 activation and expression. Our findings shed light on how Piezo1 function and expression are regulated by the substrate stiffness and highlight its involvement in the environment-mediated modulation of CFs mechanosensitivity.


Asunto(s)
Fibroblastos , Canales Iónicos , Mecanotransducción Celular , Proteínas de la Membrana , Animales , Canales Iónicos/metabolismo , Ratas , Fibroblastos/metabolismo , Fibroblastos/citología , Células Cultivadas , Calcio/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/fisiología , Miocardio/metabolismo , Miocardio/citología , Microambiente Celular
9.
Exp Biol Med (Maywood) ; 249: 10112, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38715976

RESUMEN

Chronic inflammation is a key element in the progression of essential hypertension (EH). Calcium plays a key role in inflammation, so its receptor, the calcium-sensing receptor (CaSR), is an essential mediator of the inflammatory process. Compelling evidence suggests that CaSR mediates inflammation in tissues and immune cells, where it mediates their activity and chemotaxis. Macrophages (Mφs) play a major role in the inflammatory response process. This study provided convincing evidence that R568, a positive regulator of CaSR, was effective in lowering blood pressure in spontaneously hypertensive rats (SHRs), improving cardiac function by alleviating cardiac hypertrophy and fibrosis. R568 can increase the content of CaSR and M2 macrophages (M2Mφs, exert an anti-inflammatory effect) in myocardial tissue, reduce M1 macrophages (M1Mφs), which have a pro-inflammatory effect in this process. In contrast, NPS2143, a negative state regulator of CaSR, exerted the opposite effect in all of the above experiments. Following this study, R568 increased CaSR content in SHR myocardial tissue, lowered blood pressure, promoted macrophages to M2Mφs and improved myocardial fibrosis, but interestingly, both M1Mφs and M2Mφs were increased in the peritoneal cavity of SHRs, the number of M2Mφs remained lower than M1Mφs. In vitro, R568 increased CaSR content in RAW264.7 cells (a macrophage cell line), regulating intracellular Ca2+ ([Ca2+]i) inhibited NOD-like receptor family protein 3 (NLRP3) inflammasome activation and ultimately prevented its conversion to M1Mφs. The results showed that a decrease in CaSR in hypertensive rats causes further development of hypertension and cardiac damage. EH myocardial remodeling can be improved by CaSR overexpression by suppressing NLRP3 inflammasome activation and macrophage polarization toward M1Mφs and increasing M2Mφs.


Asunto(s)
Macrófagos , Ratas Endogámicas SHR , Receptores Sensibles al Calcio , Animales , Receptores Sensibles al Calcio/metabolismo , Macrófagos/metabolismo , Ratas , Masculino , Remodelación Ventricular/fisiología , Miocardio/patología , Miocardio/metabolismo , Fibrosis/metabolismo , Presión Sanguínea , Ratones , Hipertensión/metabolismo , Hipertensión/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
10.
BMC Genomics ; 25(1): 454, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38720264

RESUMEN

BACKGROUND: In response to seasonal cold and food shortage, the Xizang plateau frogs, Nanorana parkeri (Anura: Dicroglossidae), enter a reversible hypometabolic state where heart rate and oxygen consumption in skeletal muscle are strongly suppressed. However, the effect of winter hibernation on gene expression and metabolic profiling in these two tissues remains unknown. In the present study, we conducted transcriptomic and metabolomic analyses of heart and skeletal muscle from summer- and winter-collected N. parkeri to explore mechanisms involved in seasonal hibernation. RESULTS: We identified 2407 differentially expressed genes (DEGs) in heart and 2938 DEGs in skeletal muscle. Enrichment analysis showed that shared DEGs in both tissues were enriched mainly in translation and metabolic processes. Of these, the expression of genes functionally categorized as "response to stress", "defense mechanisms", or "muscle contraction" were particularly associated with hibernation. Metabolomic analysis identified 24 and 22 differentially expressed metabolites (DEMs) in myocardium and skeletal muscle, respectively. In particular, pathway analysis showed that DEMs in myocardium were involved in the pentose phosphate pathway, glycerolipid metabolism, pyruvate metabolism, citrate cycle (TCA cycle), and glycolysis/gluconeogenesis. By contrast, DEMs in skeletal muscle were mainly involved in amino acid metabolism. CONCLUSIONS: In summary, natural adaptations of myocardium and skeletal muscle in hibernating N. parkeri involved transcriptional alterations in translation, stress response, protective mechanisms, and muscle contraction processes as well as metabolic remodeling. This study provides new insights into the transcriptional and metabolic adjustments that aid winter survival of high-altitude frogs N. parkeri.


Asunto(s)
Anuros , Hibernación , Metabolómica , Músculo Esquelético , Animales , Hibernación/genética , Hibernación/fisiología , Músculo Esquelético/metabolismo , Anuros/genética , Anuros/metabolismo , Anuros/fisiología , Miocardio/metabolismo , Transcriptoma , Perfilación de la Expresión Génica , Estaciones del Año , Metaboloma , Tibet
11.
Nat Commun ; 15(1): 3834, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714741

RESUMEN

Sleep disorders increase the risk and mortality of heart disease, but the brain-heart interaction has not yet been fully elucidated. Cuproptosis is a copper-dependent type of cell death activated by the excessive accumulation of intracellular copper. Here, we showed that 16 weeks of sleep fragmentation (SF) resulted in elevated copper levels in the male mouse heart and exacerbated myocardial ischemia-reperfusion injury with increased myocardial cuproptosis and apoptosis. Mechanistically, we found that SF promotes sympathetic overactivity, increases the germination of myocardial sympathetic nerve terminals, and increases the level of norepinephrine in cardiac tissue, thereby inhibits VPS35 expression and leads to impaired ATP7A related copper transport and copper overload in cardiomyocytes. Copper overload further leads to exacerbated cuproptosis and apoptosis, and these effects can be rescued by excision of the sympathetic nerve or administration of copper chelating agent. Our study elucidates one of the molecular mechanisms by which sleep disorders aggravate myocardial injury and suggests possible targets for intervention.


Asunto(s)
Apoptosis , Cobre , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Privación de Sueño , Animales , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Masculino , Cobre/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Ratones , Privación de Sueño/fisiopatología , Privación de Sueño/metabolismo , Privación de Sueño/complicaciones , ATPasas Transportadoras de Cobre/metabolismo , ATPasas Transportadoras de Cobre/genética , Norepinefrina/metabolismo , Norepinefrina/farmacología , Miocardio/metabolismo , Miocardio/patología , Sistema Nervioso Simpático/metabolismo , Modelos Animales de Enfermedad
12.
J Am Heart Assoc ; 13(9): e034731, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38700011

RESUMEN

BACKGROUND: Cardiac damage induced by ischemic stroke, such as arrhythmia, cardiac dysfunction, and even cardiac arrest, is referred to as cerebral-cardiac syndrome (CCS). Cardiac macrophages are reported to be closely associated with stroke-induced cardiac damage. However, the role of macrophage subsets in CCS is still unclear due to their heterogeneity. Sympathetic nerves play a significant role in regulating macrophages in cardiovascular disease. However, the role of macrophage subsets and sympathetic nerves in CCS is still unclear. METHODS AND RESULTS: In this study, a middle cerebral artery occlusion mouse model was used to simulate ischemic stroke. ECG and echocardiography were used to assess cardiac function. We used Cx3cr1GFPCcr2RFP mice and NLRP3-deficient mice in combination with Smart-seq2 RNA sequencing to confirm the role of macrophage subsets in CCS. We demonstrated that ischemic stroke-induced cardiac damage is characterized by severe cardiac dysfunction and robust infiltration of monocyte-derived macrophages into the heart. Subsequently, we identified that cardiac monocyte-derived macrophages displayed a proinflammatory profile. We also observed that cardiac dysfunction was rescued in ischemic stroke mice by blocking macrophage infiltration using a CCR2 antagonist and NLRP3-deficient mice. In addition, a cardiac sympathetic nerve retrograde tracer and a sympathectomy method were used to explore the relationship between sympathetic nerves and cardiac macrophages. We found that cardiac sympathetic nerves are significantly activated after ischemic stroke, which contributes to the infiltration of monocyte-derived macrophages and subsequent cardiac dysfunction. CONCLUSIONS: Our findings suggest a potential pathogenesis of CCS involving the cardiac sympathetic nerve-monocyte-derived macrophage axis.


Asunto(s)
Modelos Animales de Enfermedad , Accidente Cerebrovascular Isquémico , Macrófagos , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Accidente Cerebrovascular Isquémico/fisiopatología , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Receptores CCR2/genética , Receptores CCR2/metabolismo , Masculino , Ratones Noqueados , Ratones , Infarto de la Arteria Cerebral Media/fisiopatología , Infarto de la Arteria Cerebral Media/patología , Sistema Nervioso Simpático/fisiopatología , Miocardio/patología , Miocardio/metabolismo , Cardiopatías/etiología , Cardiopatías/fisiopatología , Cardiopatías/patología , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Receptor 1 de Quimiocinas CX3C/deficiencia
13.
J Am Heart Assoc ; 13(9): e032172, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38700022

RESUMEN

BACKGROUND: The purpose of this study was to investigate a therapeutic approach targeting the inflammatory response and consequent remodeling from ischemic myocardial injury. METHODS AND RESULTS: Coronary thrombus aspirates were collected from patients at the time of ST-segment-elevation myocardial infarction and subjected to array-based proteome analysis. Clinically indistinguishable at myocardial infarction (MI), patients were stratified into vulnerable and resilient on the basis of 1-year left ventricular ejection fraction and death. Network analysis from coronary aspirates revealed prioritization of tumor necrosis factor-α signaling in patients with worse clinical outcomes. Infliximab, a tumor necrosis factor-α inhibitor, was infused intravenously at reperfusion in a porcine MI model to assess whether infliximab-mediated immune modulation impacts post-MI injury. At 3 days after MI (n=7), infliximab infusion increased proregenerative M2 macrophages in the myocardial border zone as quantified by immunofluorescence (24.1%±23.3% in infliximab versus 9.29%±8.7% in sham; P<0.01). Concomitantly, immunoassays of coronary sinus samples quantified lower troponin I levels (41.72±7.34 pg/mL versus 58.11±10.75 pg/mL; P<0.05) and secreted protein analysis revealed upregulation of injury-modifying interleukin-2, -4, -10, -12, and -18 cytokines in the infliximab-treated cohort. At 4 weeks (n=12), infliximab treatment resulted in significant protective influence, improving left ventricular ejection fraction (53.9%±5.4% versus 36.2%±5.3%; P<0.001) and reducing scar size (8.31%±10.9% versus 17.41%±12.5%; P<0.05). CONCLUSIONS: Profiling of coronary thrombus aspirates in patients with ST-segment-elevation MI revealed highest association for tumor necrosis factor-α in injury risk. Infliximab-mediated immune modulation offers an actionable pathway to alter MI-induced inflammatory response, preserving contractility and limiting adverse structural remodeling.


Asunto(s)
Modelos Animales de Enfermedad , Infliximab , Remodelación Ventricular , Infliximab/uso terapéutico , Infliximab/farmacología , Animales , Humanos , Masculino , Persona de Mediana Edad , Remodelación Ventricular/efectos de los fármacos , Femenino , Infarto del Miocardio con Elevación del ST/tratamiento farmacológico , Infarto del Miocardio con Elevación del ST/inmunología , Función Ventricular Izquierda/efectos de los fármacos , Porcinos , Anciano , Factor de Necrosis Tumoral alfa/metabolismo , Volumen Sistólico/efectos de los fármacos , Trombosis Coronaria/prevención & control , Trombosis Coronaria/tratamiento farmacológico , Miocardio/patología , Miocardio/metabolismo , Miocardio/inmunología , Troponina I/sangre , Troponina I/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo
14.
J Nanobiotechnology ; 22(1): 223, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702815

RESUMEN

Cardiac muscle targeting is a notoriously difficult task. Although various nanoparticle (NP) and adeno-associated viral (AAV) strategies with heart tissue tropism have been developed, their performance remains suboptimal. Significant off-target accumulation of i.v.-delivered pharmacotherapies has thwarted development of disease-modifying cardiac treatments, such as gene transfer and gene editing, that may address both rare and highly prevalent cardiomyopathies and their complications. Here, we present an intriguing discovery: cargo-less, safe poly (lactic-co-glycolic acid) particles that drastically improve heart delivery of AAVs and NPs. Our lead formulation is referred to as ePL (enhancer polymer). We show that ePL increases selectivity of AAVs and virus-like NPs (VLNPs) to the heart and de-targets them from the liver. Serotypes known to have high (AAVrh.74) and low (AAV1) heart tissue tropisms were tested with and without ePL. We demonstrate up to an order of magnitude increase in heart-to-liver accumulation ratios in ePL-injected mice. We also show that ePL exhibits AAV/NP-independent mechanisms of action, increasing glucose uptake in the heart, increasing cardiac protein glycosylation, reducing AAV neutralizing antibodies, and delaying blood clearance of AAV/NPs. Current approaches utilizing AAVs or NPs are fraught with challenges related to the low transduction of cardiomyocytes and life-threatening immune responses; our study introduces an exciting possibility to direct these modalities to the heart at reduced i.v. doses and, thus, has an unprecedented impact on drug delivery and gene therapy. Based on our current data, the ePL system is potentially compatible with any therapeutic modality, opening a possibility of cardiac targeting with numerous pharmacological approaches.


Asunto(s)
Dependovirus , Vectores Genéticos , Miocardio , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Dependovirus/genética , Animales , Nanopartículas/química , Ratones , Miocardio/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Humanos , Ratones Endogámicos C57BL , Corazón , Terapia Genética/métodos , Técnicas de Transferencia de Gen , Hígado/metabolismo , Tropismo Viral , Células HEK293
15.
Cell Death Dis ; 15(5): 308, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693114

RESUMEN

Heart disease involves irreversible myocardial injury that leads to high morbidity and mortality rates. Numerous cell-based cardiac in vitro models have been proposed as complementary approaches to non-clinical animal research. However, most of these approaches struggle to accurately replicate adult human heart conditions, such as myocardial infarction and ventricular remodeling pathology. The intricate interplay between various cell types within the adult heart, including cardiomyocytes, fibroblasts, and endothelial cells, contributes to the complexity of most heart diseases. Consequently, the mechanisms behind heart disease induction cannot be attributed to a single-cell type. Thus, the use of multi-cellular models becomes essential for creating clinically relevant in vitro cell models. This study focuses on generating self-organizing heart organoids (HOs) using human-induced pluripotent stem cells (hiPSCs). These organoids consist of cardiomyocytes, fibroblasts, and endothelial cells, mimicking the cellular composition of the human heart. The multi-cellular composition of HOs was confirmed through various techniques, including immunohistochemistry, flow cytometry, q-PCR, and single-cell RNA sequencing. Subsequently, HOs were subjected to hypoxia-induced ischemia and ischemia-reperfusion (IR) injuries within controlled culture conditions. The resulting phenotypes resembled those of acute myocardial infarction (AMI), characterized by cardiac cell death, biomarker secretion, functional deficits, alterations in calcium ion handling, and changes in beating properties. Additionally, the HOs subjected to IR efficiently exhibited cardiac fibrosis, displaying collagen deposition, disrupted calcium ion handling, and electrophysiological anomalies that emulate heart disease. These findings hold significant implications for the advancement of in vivo-like 3D heart and disease modeling. These disease models present a promising alternative to animal experimentation for studying cardiac diseases, and they also serve as a platform for drug screening to identify potential therapeutic targets.


Asunto(s)
Fibrosis , Células Madre Pluripotentes Inducidas , Infarto del Miocardio , Miocitos Cardíacos , Organoides , Humanos , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Organoides/metabolismo , Organoides/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miocardio/patología , Miocardio/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología
16.
Sci Rep ; 14(1): 9991, 2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693202

RESUMEN

Endothelial cells (ECs) have essential roles in cardiac tissue repair after myocardial infarction (MI). To establish stage-specific and long-term effects of the ischemic injury on cardiac ECs, we analyzed their transcriptome at landmark time points after MI in mice. We found that early EC response at Day 2 post-MI centered on metabolic changes, acquisition of proinflammatory phenotypes, initiation of the S phase of cell cycle, and activation of stress-response pathways, followed by progression to mitosis (M/G2 phase) and acquisition of proangiogenic and mesenchymal properties during scar formation at Day 7. In contrast, genes involved in vascular physiology and maintenance of vascular tone were suppressed. Importantly, ECs did not return to pre-injury phenotypes after repair has been completed but maintained inflammatory, fibrotic and thrombotic characteristics and lost circadian rhythmicity. We discovered that the highest induced transcript is the mammalian-specific Sh2d5 gene that promoted migration and invasion of ECs through Rac1 GTPase. Our results revealed a synchronized, temporal activation of disease phenotypes, metabolic pathways, and proliferation in quiescent ECs after MI, indicating that precisely-timed interventions are necessary to optimize cardiac tissue repair and improve outcomes. Furthermore, long-term effects of acute ischemic injury on ECs may contribute to vascular dysfunction and development of heart failure.


Asunto(s)
Células Endoteliales , Perfilación de la Expresión Génica , Infarto del Miocardio , Animales , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Ratones , Células Endoteliales/metabolismo , Células Endoteliales/patología , Transcriptoma , Masculino , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética , Modelos Animales de Enfermedad , Proliferación Celular , Movimiento Celular/genética
17.
PLoS One ; 19(5): e0299557, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38718072

RESUMEN

The continued development in methylome analysis has enabled a more precise assessment of DNA methylation, but treatment of target tissue prior to analysis may affect DNA analysis. Prediction of age based on methylation levels in the genome (DNAmAge) has gained much interest in disease predisposition (biological age estimation), but also in chronological donor age estimation in crime case samples. Various epigenetic clocks were designed to predict the age. However, it remains unknown how the storage of the tissues affects the DNAmAge estimation. In this study, we investigated the storage method impact of DNAmAge by the comparing the DNAmAge of the two commonly used storage methods, freezing and formalin-fixation and paraffin-embedding (FFPE) to DNAmAge of fresh tissue. This was carried out by comparing paired heart tissue samples of fresh tissue, samples stored by freezing and FFPE to chronological age and whole blood samples from the same individuals. Illumina EPIC beadchip array was used for methylation analysis and the DNAmAge was evaluated with the following epigenetic clocks: Horvath, Hannum, Levine, Horvath skin+blood clock (Horvath2), PedBE, Wu, BLUP, EN, and TL. We observed differences in DNAmAge among the storage conditions. FFPE samples showed a lower DNAmAge compared to that of frozen and fresh samples. Additionally, the DNAmAge of the heart tissue was lower than that of the whole blood and the chronological age. This highlights caution when evaluating DNAmAge for FFPE samples as the results were underestimated compared with fresh and frozen tissue samples. Furthermore, the study also emphasizes the need for a DNAmAge model based on heart tissue samples for an accurate age estimation.


Asunto(s)
Metilación de ADN , Formaldehído , Miocardio , Adhesión en Parafina , Fijación del Tejido , Humanos , Adhesión en Parafina/métodos , Formaldehído/química , Miocardio/metabolismo , Fijación del Tejido/métodos , Masculino , Adulto , Femenino , Persona de Mediana Edad , Criopreservación/métodos , Adolescente , Anciano , Adulto Joven
18.
J Cell Mol Med ; 28(10): e18324, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38760897

RESUMEN

Early research suggested that bone morphogenetic protein 10 (BMP10) is primarily involved in cardiac development and congenital heart disease processes. BMP10 is a newly identified cardiac-specific protein. In recent years, reports have emphasized the effects of BMP10 on myocardial apoptosis, fibrosis and immune response, as well as its synergistic effects with BMP9 in vascular endothelium and role in endothelial dysfunction. We believe that concentrating on this aspect of the study will enhance our knowledge of the pathogenesis of diabetes and the cardiovascular field. However, there have been no reports of any reviews discussing the role of BMP10 in diabetes and cardiovascular disease. In addition, the exact pathogenesis of diabetic cardiomyopathy is not fully understood, including myocardial energy metabolism disorders, microvascular changes, abnormal apoptosis of cardiomyocytes, collagen structural changes and myocardial fibrosis, all of which cause cardiac function impairment directly or indirectly and interact with one another. This review summarizes the research results of BMP10 in cardiac development, endothelial function and cardiovascular disease in an effort to generate new ideas for future research into diabetic cardiomyopathy.


Asunto(s)
Proteínas Morfogenéticas Óseas , Enfermedades Cardiovasculares , Diabetes Mellitus , Cardiomiopatías Diabéticas , Humanos , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Apoptosis
19.
Mol Med Rep ; 30(1)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38785153

RESUMEN

17ß­estradiol (E2) can inhibit cardiac fibrosis in female patients with heart failure (HF) and activate cell division cycle 42 (Cdc42), however it is unknown whether 17ß­estradiol (E2) can ameliorate differentiation and collagen synthesis in TGF­ß1­stimulated mouse cardiac fibroblasts (MCFs) by regulating cell division cycle 42 (Cdc42). The present study aimed to investigate the roles of estrogen and Cdc42 in preventing myocardial fibrosis and the underlying molecular mechanisms. An ELISA was used to measure the levels of E2 and Cdc42 in the serum of patients with heart failure (HF), and western blotting was used to measure the expression levels of Cdc42 in TGF­ß1­stimulated immortalized MCFs. MCFs were transfected with a Cdc42 overexpression (OE) lentivirus or small interfering RNA (siRNA), or treated with a Cdc42 inhibitor (MLS­573151), and the function of Cdc42 was assessed by western blotting, immunofluorescence staining, reverse transcription­quantitative PCR and dual­luciferase reporter assays. Western blotting and immunofluorescence staining were performed to verify the protective effect of E2 on TGF­ß1­stimulated MCFs, and the association between the protective effect and Cdc42. The results demonstrated that Cdc42 levels were increased in the serum of patients with HF and were positively correlated with the levels of E2; however, Cdc42 levels were decreased in TGF­ß1­stimulated MCFs. Cdc42 inhibited MCF differentiation and collagen synthesis, as indicated by the protein expression of α­smooth muscle actin, collagen I and collagen III. Mechanistically, Cdc42 inhibited the transcription of TGF­ß1 by promoting the expression of p21 (RAC1)­activated kinase 1 (Pak1)/JNK/c­Jun signaling pathway proteins and inhibiting the activity of the Tgfb1 gene promoter. In addition, E2 inhibited the differentiation and collagen synthesis of TGF­ß1­stimulated MCFs, and promoted the protein expression of Pak1, JNK and c­Jun, consistent with the effects of Cdc42, whereas the effects of E2 were abolished when Cdc42 was knocked down. The aforementioned findings suggested that E2 could inhibit differentiation and collagen synthesis in TGF­ß1­stimulated MCFs by regulating Cdc42 and the downstream Pak1/JNK/c­Jun signaling pathway.


Asunto(s)
Diferenciación Celular , Colágeno , Estradiol , Estrógenos , Fibroblastos , Factor de Crecimiento Transformador beta1 , Proteína de Unión al GTP cdc42 , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP cdc42/genética , Animales , Diferenciación Celular/efectos de los fármacos , Ratones , Factor de Crecimiento Transformador beta1/metabolismo , Humanos , Colágeno/metabolismo , Colágeno/biosíntesis , Femenino , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Estrógenos/farmacología , Estradiol/farmacología , Persona de Mediana Edad , Miocardio/metabolismo , Insuficiencia Cardíaca/metabolismo , Masculino , Transducción de Señal/efectos de los fármacos
20.
Cell Mol Life Sci ; 81(1): 228, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38777955

RESUMEN

Diabetic cardiomyopathy (DCM) is a prevalent complication of type 2 diabetes (T2D). 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) is a glycolysis regulator. However, the potential effects of PFKFB3 in the DCM remain unclear. In comparison to db/m mice, PFKFB3 levels decreased in the hearts of db/db mice. Cardiac-specific PFKFB3 overexpression inhibited myocardial oxidative stress and cardiomyocyte apoptosis, suppressed mitochondrial fragmentation, and partly restored mitochondrial function in db/db mice. Moreover, PFKFB3 overexpression stimulated glycolysis. Interestingly, based on the inhibition of glycolysis, PFKFB3 overexpression still suppressed oxidative stress and apoptosis of cardiomyocytes in vitro, which indicated that PFKFB3 overexpression could alleviate DCM independent of glycolysis. Using mass spectrometry combined with co-immunoprecipitation, we identified optic atrophy 1 (OPA1) interacting with PFKFB3. In db/db mice, the knockdown of OPA1 receded the effects of PFKFB3 overexpression in alleviating cardiac remodeling and dysfunction. Mechanistically, PFKFB3 stabilized OPA1 expression by promoting E3 ligase NEDD4L-mediated atypical K6-linked polyubiquitination and thus prevented the degradation of OPA1 by the proteasomal pathway. Our study indicates that PFKFB3/OPA1 could be potential therapeutic targets for DCM.


Asunto(s)
Cardiomiopatías Diabéticas , GTP Fosfohidrolasas , Miocitos Cardíacos , Fosfofructoquinasa-2 , Ubiquitinación , Fosfofructoquinasa-2/metabolismo , Fosfofructoquinasa-2/genética , Animales , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/genética , Ratones , GTP Fosfohidrolasas/metabolismo , GTP Fosfohidrolasas/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Masculino , Estrés Oxidativo , Apoptosis/genética , Miocardio/metabolismo , Miocardio/patología , Ratones Endogámicos C57BL , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/genética , Glucólisis , Humanos , Estabilidad Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA