Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Mol Med (Berl) ; 102(7): 887-897, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38733386

RESUMEN

Atherosclerosis (AS) is a chronic inflammatory arterial disease, in which abnormal lipid metabolism and foam cell formation play key roles. Histamine is a vital biogenic amine catalyzed by histidine decarboxylase (HDC) from L-histidine. Histamine H1 receptor (H1R) antagonist is a commonly encountered anti-allergic agent in the clinic. However, the role and mechanism of H1R in atherosclerosis have not been fully elucidated. Here, we explored the effect of H1R on atherosclerosis using Apolipoprotein E-knockout (ApoE-/-) mice with astemizole (AST, a long-acting H1R antagonist) treatment. The results showed that AST increased atherosclerotic plaque area and hepatic lipid accumulation in mice. The result of microarray study identified a significant change of endothelial lipase (LIPG) in CD11b+ myeloid cells derived from HDC-knockout (HDC-/-) mice compared to WT mice. Blocking H1R promoted the formation of foam cells from bone marrow-derived macrophages (BMDMs) of mice by up-regulating p38 mitogen-activated protein kinase (p38 MAPK) and LIPG signaling pathway. Taken together, these findings demonstrate that blocking H1R signal aggravates atherosclerosis by promoting abnormal lipid metabolism and macrophage-derived foam cell formation via p38 MAPK-LIPG signaling pathway. KEY MESSAGES: Blocking H1R signal with AST aggravated atherosclerosis and increased hepatic lipid accumulation in high-fat diet (HFD)-fed ApoE-/- mice. Blocking H1R signal promoted macrophage-derived foam cell formation via p38 MAPK-LIPG signaling pathway.


Asunto(s)
Aterosclerosis , Células Espumosas , Inflamación , Ratones Noqueados , Receptores Histamínicos H1 , Transducción de Señal , Animales , Células Espumosas/metabolismo , Células Espumosas/patología , Aterosclerosis/metabolismo , Aterosclerosis/etiología , Aterosclerosis/patología , Ratones , Inflamación/metabolismo , Inflamación/patología , Receptores Histamínicos H1/metabolismo , Receptores Histamínicos H1/genética , Masculino , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Ratones Endogámicos C57BL , Metabolismo de los Lípidos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Modelos Animales de Enfermedad , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Dieta Alta en Grasa/efectos adversos
2.
Int Immunopharmacol ; 123: 110774, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37567012

RESUMEN

Periodontal disease is a chronic inflammatory disease that is highly correlated with cardiovascular disease(CVD). Histamine has been proven to participate in the pathophysiological processes of cardiovascular disease and oral inflammation. However, the role of histamine in the development of cardiac microthrombosis caused by periodontal disease has not been fully elucidated. We established a murine periodontal inflammation model by injecting lipopolysaccharide (LPS) or Porphyromonas gingivalis (P. gingivalis). In order to examine the effect of histamine/H1R signaling on cardiac injury after periodontal disease, we used histidine decarboxylase- knockout (HDC-/-) mice and histamine 1 receptor (H1R) antagonist. Our results demonstrated that LPS-induced periodontal inflammation significantly increased CD11b+Gr-1+ neutrophils in the peripheral blood and myocardial interstitium. Histamine deficiency resulted in further increases in P. gingivalis, neutrophils, inflammatory cytokines, and cardiac microthrombosis in the myocardium of HDC-/- mice compared to wild-type (WT) mice. Mechanistic analysis showed that blocking H1R could synergistically interact with LPS, further increasing the phosphorylation of p65, exacerbating the inflammatory response of neutrophils and endothelial cell damage. Conclusively, the disruption of histamine-H1R signaling exacerbates cardiac microthrombosis after periodontal disease via TLR4/NFκB-p65 pathway. Our findings not only reveal a link between periodontal inflammation and myocardial injury but also provided some thoughts for the use of H1R antagonist in clinical practice.


Asunto(s)
Enfermedades Cardiovasculares , Enfermedades Periodontales , Animales , Ratones , Histamina/metabolismo , Antagonistas de los Receptores Histamínicos H1 , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Receptores Histamínicos H1/metabolismo , Receptor Toll-Like 4/metabolismo , Receptores Histamínicos
3.
Cell Biol Int ; 47(9): 1573-1588, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37303238

RESUMEN

Intimal thickening caused by the excessive multiplication of vascular smooth muscle cells (VSMCs) is the pathological process central to cardiovascular diseases, including restenosis. In response to vascular injury, VSMCs would undergo phenotypic switching from a fully differentiated, low proliferative rate phenotype to a more pro-proliferative, promigratory, and incompletely-differentiated state. The lack of a full understanding of the molecular pathways coupling the vascular injury stimuli to VSMCs phenotype switching largely limits the development of medical therapies for treating intima hyperplasia-related diseases. The role of signal transducers and activators of transcription 6 (STAT6) in modulating the proliferation and differentiation of various cell types, especially macrophage, has been well investigated, but little is known about its pathophysiological role and target genes in restenosis after vascular injury. In the present work, Stat6-/- mice were observed to exhibit less severe intimal hyperplasia compared with Stat6+/+ mice after carotid injury. The expression of STAT6 was upregulated in VSMCs located in the injured vascular walls. STAT6 deletion leads to decreased proliferation and migration of VSMCs while STAT6 overexpression enhances the proliferation and migration of VSMCs companies with reduced expression of VSMCs marker genes and organized stress fibers. The effect of STAT6 in mouse VSMCs was conserved in human aortic SMCs. RNA-deep-sequencing and experiments verification revealed LncRNA C7orf69/LOC100996318-miR-370-3p/FOXO1-ER stress signaling as the downstream network mediating the pro-dedifferentiation effect of STAT6 in VSMCs. These findings broaden our understanding of vascular pathological molecules and throw a beam of light on the therapy of a variety of proliferative vascular diseases.


Asunto(s)
Miocitos del Músculo Liso , Neointima , Factor de Transcripción STAT6 , Animales , Ratones , Factor de Transcripción STAT6/metabolismo , Miocitos del Músculo Liso/citología , Músculo Liso Vascular/citología , Neointima/patología , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología , Ratones Noqueados , Humanos , Ratones Endogámicos BALB C , Masculino , Femenino , Persona de Mediana Edad , Anciano , Células Cultivadas , Aorta/citología , Desdiferenciación Celular
4.
Free Radic Biol Med ; 192: 98-114, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36165929

RESUMEN

Doxorubicin (DOX) is widely used in the treatment of various cancers, increasing the great risk of adverse cardiovascular events, while the clinical intervention effect is not ideal. Histamine has been documented to participate in pathophysiological processes of cardiovascular diseases and inflammation-associated carcinogenesis. However, the potential roles of histamine in antitumor-related cardiotoxicity have not been fully elucidated. In this study, cardiomyocytes (hiPSC-CMs, HL-1 cells) and mice were treated with DOX to establish DOX-induced cardiotoxicity (DIC) models. Histidine decarboxylase knockout mice (HDC-/-) mice and histamine 1 receptor (H1R) antagonist were used to explore the effect of histamine/H1R signaling on DIC. Our results demonstrated that histamine deficiency or pharmaceutical inhibition of H1R accelerated myocardial ferroptosis, which is responsible for the aggravated DIC both in vivo and in vitro, while the supplementation of exogenous histamine reversed these changes. Our data revealed that the dysfunction of histamine/H1R signaling repressed the activation of transducer and activator of transcription 3 (STAT3), accompanying with decreased expression of solute carrier family7member11 (SLC7A11), a major modulator of ferroptosis. Conclusively, the disruption of histamine/H1R axis triggered ferroptosis and exacerbated DIC possibly by modulating STAT3-SLC7A11 pathway. Our findings point to a potential therapeutic target for DIC and provide more consideration on the usage of antihistamine drugs.


Asunto(s)
Ferroptosis , Histamina , Animales , Cardiotoxicidad/genética , Doxorrubicina/metabolismo , Doxorrubicina/toxicidad , Ferroptosis/genética , Histamina/metabolismo , Histamina/farmacología , Antagonistas de los Receptores Histamínicos/metabolismo , Antagonistas de los Receptores Histamínicos/farmacología , Antagonistas de los Receptores Histamínicos H1/farmacología , Histidina Descarboxilasa/metabolismo , Histidina Descarboxilasa/farmacología , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Preparaciones Farmacéuticas/metabolismo
5.
Acta Pharm Sin B ; 12(4): 1840-1855, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35847488

RESUMEN

Neutrophils are mobilized and recruited to the injured heart after myocardial infarction, and neutrophil count has been clinically implicated to be associated with coronary disease severity. Histidine decarboxylase (HDC) has been implicated in regulating reactive oxidative species (ROS) and the differentiation of myeloid cells. However, the effect of HDC on neutrophils after myocardial infarction remains unclear. Here, we found that neutrophils were disorderly recruited into the ischemic injured area of the myocardium of Hdc deficiency (Hdc -/-) mice. Moreover, Hdc deficiency led to attenuated adhesion but enhanced migration and augmented ROS/neutrophil extracellular traps (NETs) production in neutrophils. Hdc -/- mouse-derived NETs promoted cardiomyocyte death and cardiac fibroblast proliferation/migration. Furthermore, protein arginine methyltransferase 1 (PRMT1) was increased in Hdc -/- mouse-derived neutrophils but decreased with exogenous histamine treatment. Its expression could be rescued by blocking histamine receptor 1 (H1R), inhibiting ATP synthesis or reducing SWItch/sucrose non fermentable (SWI/SNF) chromatin remodeling complex. Accordingly, histamine or MS023 treatment could decrease ROS and NETs ex vivo, and ameliorated cardiac function and fibrosis, along with the reduced NETs in plasma in vivo. Together, our findings unveil the role of HDC in NETosis by histamine-H1R-ATP-SWI/SNF-PRMT1-ROS signaling and provide new biomarkers and targets for identifying and tuning the detrimental immune state in cardiovascular disease.

6.
Biochem Biophys Res Commun ; 588: 161-167, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34954523

RESUMEN

Distinct macrophage populations exert highly heterogeneity and perform various functions, among which, a crucial function of lipid metabolism is highlighted. However, the role of histidine metabolism disorder in macrophage lipid metabolism remains elusive. Addressed this question, we sorted and cultured the bone marrow-derived macrophages (BMDMs) of histidine decarboxylase (Hdc) knockout (Hdc-/-) mice with an in vitro oxidized low-density lipoprotein (ox-LDL) model, and detected the intracellular lipids by Oil Red O staining as well as lipid probe staining. Astemizole, a canonical and long-acting histamine H1 receptor (H1R) antagonist, was applied to elucidate the impact of antagonizing the H1R-dependent signaling pathway on macrophage lipid metabolism. Subsequently, the differential expressed genes were screened and analyzed in the bone marrow-derived CD11b+ immature myeloid cells of Hdc-/- and Hdc+/+ mice with a high fat diet by the microarray study. The expression levels of cholesterol metabolism-related genes were examined by qRT-PCR to explore underlying mechanisms. Lastly, we used a high-sensitivity histidine probe to detect the intracellular histidine in the BMDMs after oxidative stress. The results revealed that histidine metabolism disorder and histamine deficiency aggravated lipid accumulation in the ox-LDL-treated BMDMs. The expression level of H1R gene in the BMDMs was down-regulated after ox-LDL stimulation. The disruption of the H1R-dependent signaling pathway by astemizole further exacerbated ox-LDL-induced lipid deposition in the BMDMs partly by up-regulating scavenger receptor class A (SR-A) for lipid intake, down-regulating neutral cholesteryl ester hydrolase (nCEH) for cholesterol esterification and down-regulating ATP-binding cassette transporters A1 (ABCA1) and ABCG1 for reverse cholesterol transport. The intracellular histidine increased under ox-LDL condition, which was further increased by Hdc knockout. Collectively, these results partially reveal the relationship between histidine metabolism and lipid metabolism in the BMDMs and offer a novel strategy for lipid metabolism disorder-associated diseases.


Asunto(s)
Histidina/metabolismo , Metabolismo de los Lípidos , Lipoproteínas LDL/farmacología , Macrófagos/metabolismo , Animales , Colesterol/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Histamina/deficiencia , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Histamínicos/metabolismo , Transducción de Señal/efectos de los fármacos
7.
J Cardiovasc Transl Res ; 15(3): 621-634, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34734351

RESUMEN

Myocardial infarction (MI) is a significant contributor to the development of heart failure. Histidine decarboxylase (HDC), the unique enzyme that converts L-histidine to histamine, is highly expressed in CD11b+ immature myeloid cells. However, the relationship between HDC-expressing macrophages and cardiac myofibroblasts remains to be explained. Here, we demonstrate that the GFP (green fluorescent protein)-labeled HDC+CD11b+ myeloid precursors and their descendants could differentiate into fibroblast-like cells in myocardial interstitium. Furthermore, we prove that CD11b+Ly6C+ monocytes/macrophages, but not CD11b+Ly6G+ granulocytes, are identified as the main cellular source for bone marrow-derived myofibroblast transformation, which could be regulated via histamine H1 and H2 receptor-dependent signaling pathways. Using HDC knockout mice, we find that histamine deficiency promotes myofibroblast transformation from Ly6C+ macrophages and cardiac fibrosis partly through upregulating the expression of Krüppel-like factor 5 (KLF5). Taken together, our data uncover a central role of HDC in regulating bone marrow-derived macrophage-to-myofibroblast transformation but also identify a histamine receptor (HR)-KLF5 related signaling pathway that mediates myocardial fibrosis post-MI. CD11b+Ly6C+ monocytes/macrophages are the main cellular source for bone marrow-derived myofibroblast transformation. Histamine inhibits myofibroblasts transformation via H1R and H2R-dependent signaling pathways, and ameliorates cardiac fibrosis partly through upregulating KLF5 expression.


Asunto(s)
Histamina , Histidina Descarboxilasa , Células Mieloides , Infarto del Miocardio , Miofibroblastos , Animales , Fibrosis , Histamina/deficiencia , Histidina Descarboxilasa/metabolismo , Ratones , Células Mieloides/citología , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Miofibroblastos/citología
8.
Int J Mol Sci ; 22(14)2021 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-34299019

RESUMEN

Myocardial infarction (MI) is one of the most common cardiovascular diseases. Although previous studies have shown that histidine decarboxylase (HDC), a histamine-synthesizing enzyme, is involved in the stress response and heart remodeling after MI, the mechanism underlying it remains unclear. In this study, using Hdc-deficient mice (Hdc-/- mice), we established an acute myocardial infarction mouse model to explore the potential roles of Hdc/histamine in cardiac immune responses. Comprehensive analysis was performed on the transcriptomes of infarcted hearts. Differentially expressed gene (DEG) analysis identified 2126 DEGs in Hdc-deficient groups and 1013 in histamine-treated groups. Immune related pathways were enriched in Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Then we used the ssGSEA algorithm to evaluate 22 kinds of infiltrated immunocytes, which indicated that myeloid cells and T memory/follicular helper cells were tightly regulated by Hdc/histamine post MI. The relationships of lncRNAs and the Gene Ontology (GO) functions of protein-coding RNAs and immunocytes were dissected in networks to unveil immune-associated lncRNAs and their roles in immune modulation after MI. Finally, we screened out and verified four lncRNAs, which were closely implicated in tuning the immune responses after MI, including ENSMUST00000191157, ENSMUST00000180693 (PTPRE-AS1), and ENSMUST-00000182785. Our study highlighted the HDC-regulated myeloid cells as a driving force contributing to the government of transmission from innate immunocytes to adaptive immunocytes in the progression of the injury response after MI. We identified the potential role of the Hdc/histamine-lncRNAs network in regulating cardiac immune responses, which may provide novel promising therapeutic targets for further promoting the treatment of ischemic heart disease.


Asunto(s)
Histidina Descarboxilasa/metabolismo , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , ARN Largo no Codificante/metabolismo , Transcriptoma/genética , Algoritmos , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Ontología de Genes , Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/inmunología , Histidina Descarboxilasa/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células Mieloides/inmunología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Células RAW 264.7 , ARN Largo no Codificante/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T Colaboradores-Inductores/inmunología
9.
Onco Targets Ther ; 14: 2241-2246, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33833523

RESUMEN

Bronchiolar adenoma (BA) is recognized as a neoplasm with benign clinical course. Histologically, BA is characterized by nodular proliferation of the bilayered bronchiolar-type epithelium, including multipartite epithelial cells and a continuous layer of basal cells. Recent reports have revealed the frequent presence of driver gene mutations in BA, suggesting its neoplastic nature. However, it is still debatable whether BA has malignant potential. Herein, we report the first case of BA harboring the same KRAS mutation with the adjacent invasive mucinous adenocarcinoma (IMA). Additionally, the loss of continuity of the basal cell layer in the junctional zone between BA and IMA indicated a malignant transformation from BA to IMA in this particular case.

10.
Front Physiol ; 11: 579712, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33192584

RESUMEN

Cardiac fibrosis is an important pathological basis of various cardiovascular diseases. The roles of STAT6 signal in allergy, immune regulation, tumorigenesis, and renal fibrosis have been documented. However, the function and mechanism of STAT6 signal in sympathetic overactivation-induced cardiac fibrosis have not been fully elucidated. This study explores the novel role of STAT6 signal in isoproterenol (ISO)-induced cardiac fibrosis through the regulation of inflammatory response and the differentiation of macrophages from immature myeloid cells. The expression levels of STAT6, ß1-adrenergic receptor (ß1-AR), and inflammatory factors [interleukin α (IL-1α), IL-6, IL-18, and transforming growth factor ß (TGF-ß)] in CD11b+ myeloid cells were analyzed with a microarray study. The levels of IL-6 and TGF-ß1 in the CD11b+ myeloid cells-derived macrophages were detected with reverse transcriptase-polymerase chain reaction (RT-PCR). STAT6-knockout (KO) and WT mice were used to establish a murine cardiac fibrosis model by ISO injection. Cardiac fibroblasts were isolated from the hearts of newborn STAT6-KO and WT mice, and STAT6 expression was measured by Western blotting and RT-PCR after ISO stimulation, while α-smooth muscle actin (α-SMA) expression was detected by immunofluorescence and immunohistochemistry staining. Cardiac function and pathological characteristics were examined by echocardiography and immunohistochemistry staining, respectively. Immunohistochemistry staining with anti-CD11b was performed to detect the infiltration of CD11b+ myeloid cells in heart tissue. Flow cytometry analysis was used to measure the percentages of CD11b+ myeloid cells and CD11b+Ly6C+ macrophages in the peripheral blood. The results showed that STAT6 was highly expressed in CD11b+ myeloid cells located in injured hearts, and STAT6 expression in cardiac fibroblasts was down-regulated after ISO treatment. STAT6 deficiency further aggravated ISO-induced increased expression of α-SMA in cardiac fibroblasts, myocardial fibrosis, and cardiac dysfunction. The activation of ISO/ß1-AR signal aggravated cardiac inflammatory infiltration, promoted CD11b+ myeloid cell mobilization, and enhanced CD11b+Ly6C+/low macrophage differentiation, which was further exacerbated by STAT6 deficiency. Furthermore, ß1-AR mRNA expression significantly increased in splenic CD11b+ myeloid cells compared to their bone marrow-derived controls, and STAT6 deficiency promoted ß1-AR expression in an MI-induced sensitive cardiac fibrosis mouse model. The spleen-derived CD11b+ myeloid cells of STAT6-KO mice produced more IL-1α, IL-18, and TGF-ß than their WT counterparts. Taken together, these results suggest that STAT6 signal plays a critical role in ISO-induced ß1-AR overactivation and systemic inflammatory cascades, contributing to cardiac fibrogenesis. STAT6 should be a promising cardioprotective target against myocardial fibrosis and heart failure after ß1-AR overactivation-induced myocardial injury.

11.
Stem Cell Res Ther ; 11(1): 27, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32127042

RESUMEN

BACKGROUND: The efficiency and quality of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are crucial for regenerative medicine, disease modeling, drug screening, and the study of the development events during cardiac specification. However, their applications have been hampered by the differentiation efficiency, poor maturation, and high interline variability. Recent studies have reported that histamine plays important roles in hematopoietic stem cell proliferation and neutrophil maturation. However, its roles in cardiovascular tissue regeneration have not been thoroughly investigated. In the current study, we identified a novel physiological function of the histamine/histamine 1 receptor (H1R) signal in regulating the differentiation of hiPSC-CMs and heart development. METHODS: Transgenic zebrafish model (cmlc2: mCherry) was treated with histamine and histamine receptor (HR) antagonists. Histological morphology and ultrastructure of zebrafish heart were measured. Histamine-deficient pregnant mice (HDC-/-) were treated with H1R antagonist (pyrilamine) by intragastric administration from E8.5 to E18.5. Cardiac histological morphology and ultrastructure were analyzed in neonatal mice, and cardiac function in adult mice was measured. In vitro, histamine and HR antagonists were administrated in the culture medium during hiPSC-CM differentiation at different stages. The efficiency and maturation of cardiac differentiation were evaluated. Finally, histamine-treated hiPSC-CMs were transplanted into ischemic myocardium to detect the possible therapeutic effect. RESULTS: Administration of H1R antagonist during heart development induced cardiac dysplasia in zebrafish. Furthermore, using histidine decarboxylase (HDC) knockout mice, we examined abnormal swelling of myocardial mitochondria and autophagy formation under the condition of endogenous histamine deficiency. Histamine significantly promoted myocardial differentiation from human induced pluripotent stem cells (hiPSCs) with better structure and function via a H1R-dependent signal. The activation of histamine/H1R signaling pathway augmented hiPSC-derived cardiomyocyte (hiPSC-CM) differentiation through the ERK1/2-STAT3 signaling pathway. In addition, histamine-pre-treated hiPSC-CMs were transplanted into the ischemic hearts of myocardial injured mice and exhibited better survival and myocardial protection. CONCLUSIONS: Thus, these findings indicated that histamine/H1R and its downstream signals were not only involved in cardiac differentiation but also provided a better survival environment for stem cell transplanted into ischemic myocardium.


Asunto(s)
Histamina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Miocitos Cardíacos/metabolismo , Transducción de Señal , Pez Cebra
12.
J Cell Mol Med ; 24(6): 3504-3520, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32064748

RESUMEN

Neutrophil-platelet interactions are responsible for thrombosis as well as inflammatory responses following acute myocardial infarction (AMI). While histamine has been shown to play a crucial role in many physiological and pathological processes, its effects on neutrophil-platelet interactions in thromboinflammatory complications of AMI remain elusive. In this study, we show a previously unknown mechanism by which neutrophil-derived histamine protects the infarcted heart from excessive neutrophil-platelet interactions and redundant arterial thrombosis. Using histamine-deficient (histidine decarboxylase knockout, HDC-/- ) and wild-type murine AMI models, we demonstrate that histamine deficiency increases the number of microthrombosis after AMI, in accordance with depressed cardiac function. Histamine-producing myeloid cells, mainly Ly6G+ neutrophils, directly participate in arteriole thrombosis. Histamine deficiency elevates platelet activation and aggregation by enhancing Akt phosphorylation and leads to dysfunctional characteristics in neutrophils which was confirmed by high levels of reactive oxygen species production and CD11b expression. Furthermore, HDC-/- platelets were shown to elicit neutrophil extracellular nucleosomes release, provoke neutrophil-platelet interactions and promote HDC-expressing neutrophils recruitment in arteriole thrombosis in vivo. In conclusion, we provide evidence that histamine deficiency promotes coronary microthrombosis and deteriorates cardiac function post-AMI, which is associated with the enhanced platelets/neutrophils function and neutrophil-platelet interactions.


Asunto(s)
Plaquetas/patología , Comunicación Celular , Vasos Coronarios/patología , Histamina/deficiencia , Infarto del Miocardio/complicaciones , Neutrófilos/patología , Trombosis/etiología , Animales , Plaquetas/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Vasos Coronarios/efectos de los fármacos , Histamina/farmacología , Histidina Descarboxilasa/deficiencia , Ratones , Modelos Biológicos , Infarto del Miocardio/patología , Miocardio/patología , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Histamínicos/metabolismo , Trombopoyesis/efectos de los fármacos , Trombosis/patología
13.
Front Immunol ; 10: 2638, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31798581

RESUMEN

Emerging evidence shows that signal transducer and activator of transcription 6 (STAT6) plays critical roles in tumor development. We previously found high-level expression of STAT6 in human lung adenocarcinoma and squamous cell carcinoma, specifically in infiltrated immune cells located in the lung interstitium. Nevertheless, the role of STAT6 signaling in lung carcinogenesis and lung cancer proliferation and its underlying mechanisms remain unclear. This study aimed to investigate the role of STAT6 and the interaction between STAT6 and the tumor microenvironment in pulmonary tumorigenesis. We established a murine model of primary lung carcinogenesis in STAT6-deficient (STAT6-/-) and STAT6 wild-type (WT) BALB/c mice using the carcinogen urethane. Two-month-old male mice were intraperitoneally injected with urethane (1 g/kg) dissolved in phosphate buffered saline (PBS). Primary tumors were monitored in vivo by positron emission tomography scanning. At 4, 6, and 9 months after urethane injection, lung tumors were harvested from the STAT6-/- and WT mice for analysis. Small interfering RNA was used to downregulate the expression of STAT6 in tumor cells. Fluorescence activated cell sorting analysis was used to analyze fluorescence-conjugated cell markers. Transwell assays were used in coculturing experiments. STAT6 protein expression was detected by Western blotting, immunohistochemistry, and immunofluorescence. STAT6 mRNA expression was detected by quantitative real time-polymerase chain reaction. Cell Counting Kit-8 and colony formation assays were performed to evaluate cell proliferation. We detected high expression of STAT6 in CD11b+ cells of lung carcinoma. Our results indicate that STAT6 deficiency inhibits carcinogen-induced tumor growth and improves prognosis. STAT6 deficiency also decreased the mobilization and differentiation of CD11b+ cells. STAT6 deficiency in CD11b+ cells but not tumor cells decreased interleukin (IL)-4 secretion and the differentiation of CD11b+ cells into M2 macrophage cells. In conclusion, our findings indicate that IL-4/STAT6 signaling in CD11b+ cells promotes lung cancer progression by triggering an IL-4 positive feedback loop and increasing M2 myeloid cells. STAT6 may be a new therapeutic target for the prevention and treatment of lung cancer.


Asunto(s)
Interleucina-4/fisiología , Neoplasias Pulmonares/etiología , Células Mieloides/patología , Factor de Transcripción STAT6/fisiología , Animales , Antígeno CD11b/análisis , Diferenciación Celular , Movimiento Celular , Polaridad Celular , Progresión de la Enfermedad , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/fisiología
14.
J Cell Mol Med ; 23(12): 8392-8409, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31600036

RESUMEN

Histidine decarboxylase (HDC) catalyses the formation of histamine from L-histidine. Histamine is a biogenic amine involved in many physiological and pathological processes, but its role in the regeneration of skeletal muscles has not been thoroughly clarified. Here, using a murine model of hindlimb ischaemia, we show that histamine deficiency in Hdc knockout (Hdc-/- ) mice significantly reduces blood perfusion and impairs muscle regeneration. Using Hdc-EGFP transgenic mice, we demonstrate that HDC is expressed predominately in CD11b+ Gr-1+ myeloid cells but not in skeletal muscles and endothelial cells. Large amounts of HDC-expressing CD11b+ myeloid cells are rapidly recruited to injured and inflamed muscles. Hdc-/- enhances inflammatory responses and inhibits macrophage differentiation. Mechanically, we demonstrate that histamine deficiency decreases IGF-1 (insulin-like growth factor 1) levels and diminishes myoblast proliferation via H3R/PI3K/AKT-dependent signalling. These results indicate a novel role for HDC-expressing CD11b+ myeloid cells and histamine in myoblast proliferation and skeletal muscle regeneration.


Asunto(s)
Proliferación Celular/fisiología , Histamina/deficiencia , Inflamación/fisiopatología , Músculo Esquelético/fisiopatología , Mioblastos/metabolismo , Regeneración/fisiología , Animales , Línea Celular , Proliferación Celular/genética , Histidina Descarboxilasa/deficiencia , Histidina Descarboxilasa/genética , Inflamación/genética , Inflamación/metabolismo , Isquemia/fisiopatología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/metabolismo , Mioblastos/citología , Regeneración/genética , Transducción de Señal/fisiología
15.
Oxid Med Cell Longev ; 2019: 1209307, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31249639

RESUMEN

Genetically human apolipoprotein E (APOE) ε32 is associated with a decreased risk of ischemic heart disease. ApoE deficiency in mice impairs infarct healing after myocardial infarction (MI). After the ischemic injury, a large number of neutrophils are firstly recruited into the infarct zone and then degrade dead material and promote reparative phase transformation. The role of ApoE in inflammation response in the early stage of MI remains largely unclear. In this study, we investigated the effect of ApoE deficiency on neutrophils' function and myocardial injury after myocardial infarction. By left coronary artery ligation in ApoE -/- and wild-type (WT) mice, we observed increased infarct size and neutrophil infiltration in ApoE -/- mice. Within the infarct zone, more neutrophil extracellular traps (NETs) were observed in ApoE -/- mice, while increased ex vivo NET formation was detected in ApoE -/- mouse-derived neutrophils through the NADPH oxidase-ROS-dependent pathway. Suppressing overproduced NETs reduced myocardial injury in ApoE -/- mice after ligation. In general, our findings reveal a critical role of apolipoprotein E in regulating Ly6G+ neutrophil activation and NET formation, resulting in limiting myocardial injury after myocardial infarction. In such a process, apolipoprotein E regulates NET formation via the ROS-MAPK-MSK1 pathway.


Asunto(s)
Modelos Animales de Enfermedad , Trampas Extracelulares/fisiología , Inflamación/etiología , Infarto del Miocardio/complicaciones , Daño por Reperfusión Miocárdica/etiología , Neutrófilos/patología , Especies Reactivas de Oxígeno/metabolismo , Animales , Apolipoproteínas E/deficiencia , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Infiltración Neutrófila , Neutrófilos/metabolismo
16.
Cell Death Dis ; 9(6): 694, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29880830

RESUMEN

Histamine is a widely distributed biogenic amine involved in the regulation of an array of biological processes. Serum histamine level is markedly elevated in the early stages of acute myocardial infarction, whereas the role it plays remains unclear. Histidine decarboxylase (HDC) is the unique enzyme responsible for histamine production, and cardiac injury is significantly aggravated in HDC knockout mice (HDC-/-), in which histamine is deficient. We also observed that autophagy was highly activated in cardiomyocytes of HDC-/- mice post acute myocardial infarction (AMI), which was abolished by compensation of exogenous histamine. The in vivo and in vitro results showed that acting through histamine 1 receptor, histamine increased miR-206 and miR-216b, which worked in concert to target to Atg13, resulting in the reduction of autophagy activation under hypoxia and AMI condition. Further study revealed that Atg13 interacted with FADD to promote the activation of caspase-8 and cell apoptosis. Taken together, these data unveil a novel intracellular signaling pathway involved in histamine regulating myocardial autophagy and apoptosis under hypoxia and AMI condition, which might help to more comprehensively evaluate the usage of histamine receptor antagonists and to develop new therapeutic targets for myocardial infarction.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Autofagia , Histamina/deficiencia , MicroARNs/metabolismo , Miocardio/patología , Animales , Secuencia de Bases , Caspasa 8/metabolismo , Hipoxia de la Célula , Línea Celular , Activación Enzimática , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Humanos , Ratones , MicroARNs/genética , Modelos Biológicos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/ultraestructura , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , Receptores Histamínicos/metabolismo
17.
Atherosclerosis ; 263: 42-52, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28600950

RESUMEN

BACKGROUND AND AIMS: The enzyme histidine decarboxylase (Hdc), which generates histamine, is highly expressed in CD11b+Gr-1+ myeloid cells that play a critical role in infection, inflammation and tumorigenesis. The aim of this study was to explore the role of Hdc-expressing CD11b+ myeloid cells or histamine in atherogenesis. METHODS: Hdc-EGFP bacterial artificial chromosome (BAC) transgenic reporter mice (Hdc-EGFP) were used to track Hdc expression during the development of atherosclerosis. The expression of EGFP fluorescence was examined by immunofluorescence staining in a variety of adult tissues. Wild-type (WT), Apoe knockout (Apoe-/-), Hdc knockout (Hdc-/-), and Stat6 knockout (Stat6-/-) mice were used. Serum concentration of histamine was determined with ELISA. Changes in subsets of immune cells were evaluated by flow cytometry (FACS). Non-invasive tracking of the expression of CD11b+ myeloid cells was tested using 125I-anti-CD11b SPECT/CT imaging in the early stages of atherogenesis. Microarray analysis and RT-PCR were applied to detect gene expressions while Western blot was used to assess protein levels. RESULTS: Using Hdc-EGFP transgenic mice, we demonstrated that Hdc+CD11b+ myeloid cells increase in the circulation in response to hypercholesterolemia and contribute to foam cell formation in atherosclerosis. Histamine deficiency in Hdc knockout (Hdc-/-) mice repressed the differentiation of CD11b+Ly6Chigh classically activated M1-type macrophages and CD11b+CD11c+ dendritic cells (DCs), which was associated with downregulation of signal transducer and activator of transcription 6 (Stat6) expression. Furthermore, the results of in vivo and in vitro studies demonstrated that histamine could promote macrophage differentiation and foam cell formation via the Stat6 signal. CONCLUSIONS: Modulation of histamine and Stat6-signaling may represent an attractive therapeutic strategy for the prevention or treatment of atherosclerosis.


Asunto(s)
Antígeno CD11b/metabolismo , Células Espumosas/citología , Histamina/metabolismo , Macrófagos/citología , Células Mieloides/citología , Factor de Transcripción STAT6/metabolismo , Animales , Aterosclerosis , Diferenciación Celular , Células Dendríticas/citología , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único
18.
Sci Rep ; 7: 44007, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28272448

RESUMEN

Histamine has pleiotropic pathophysiological effects, but its role in myocardial infarction (MI)-induced cardiac remodeling remains unclear. Histidine decarboxylase (HDC) is the main enzyme involved in histamine production. Here, we clarified the roles of HDC-expressing cells and histamine in heart failure post-MI using HDC-EGFP transgenic mice and HDC-knockout (HDC-/-) mice. HDC+CD11b+ myeloid cell numbers markedly increased in the injured hearts, and histamine levels were up-regulated in the circulation post-MI. HDC-/- mice exhibited more adverse cardiac remodeling, poorer left ventricular function and higher mortality by increasing cardiac fibrogenesis post-MI. In vitro assays further confirmed that histamine inhibited heart fibroblast proliferation. Furthermore, histamine enhanced the signal transducer and activator of transcription (STAT)-6 phosphorylation level in murine heart fibroblasts, and the inhibitive effects of histamine on fibroblast proliferation could be blocked by JAK3/STAT6 signaling selective antagonist. STAT6-knockout (STAT6-/-) mice had a phenotype similar to that of HDC-/- mice post-MI; however, in contrast to HDC-/- mice, the beneficial effects of exogenous histamine injections were abrogated in STAT6-/- mice. These data suggest that histamine exerts protective effects by modulating cardiac fibrosis and remodeling post-MI, in part through the STAT6-dependent signaling pathway.


Asunto(s)
Insuficiencia Cardíaca/patología , Histamina/metabolismo , Infarto del Miocardio/patología , Remodelación Ventricular/fisiología , Animales , Citocinas/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Histamina/sangre , Histamina/farmacología , Histidina Descarboxilasa/deficiencia , Histidina Descarboxilasa/genética , Humanos , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/citología , Células Mieloides/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/veterinaria , Miocardio/citología , Miocardio/patología , Fosforilación , Receptores Histamínicos/metabolismo , Factor de Transcripción STAT6/antagonistas & inhibidores , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Transducción de Señal/efectos de los fármacos
19.
Sci Rep ; 5: 13131, 2015 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-26278136

RESUMEN

Histamine is a biogenic amine that is widely distributed and has multiple functions, but the role it plays in acute myocardial infarction (AMI) remains unclear. In this study, we investigated the origin and contribution of endogenous histamine to AMI. Histidine decarboxylase (HDC) is the unique enzyme responsible for histamine generation. Using HDC-EGFP bacterial artificial chromosome (BAC) transgenic mice in which EGFP expression is controlled by the HDC promoter, we identified HDC expression primarily in CD11b(+)Gr-1(+) immature myeloid cells (IMCs) that markedly increase in the early stages of AMI. Deficiency of histamine in HDC knockout mice (HDC(-/-)) reduced cardiac function and exacerbated the injury of infarcted heart. Furthermore, administering either an H1 receptor antagonist (pyrilamine) or an H2 receptor antagonist (cimetidine) demonstrated a protective effect of histamine against myocardial injury. The results of in vivo and in vitro assays showed that histamine deficiency promotes the apoptosis of cardiomyocytes and inhibits macrophage infiltration. In conclusion, CD11b(+)Gr-1(+) IMCs are the predominant HDC-expressing sites in AMI, and histamine plays a protective role in the process of AMI through inhibition of cardiomyocyte apoptosis and facilitation of macrophage infiltration.


Asunto(s)
Apoptosis/efectos de los fármacos , Histamina/farmacología , Histidina Descarboxilasa/genética , Macrófagos/inmunología , Infarto del Miocardio/patología , Enfermedad Aguda , Animales , Línea Celular , Cimetidina/farmacología , Forma MB de la Creatina-Quinasa/análisis , Modelos Animales de Enfermedad , Lesiones Cardíacas/etiología , Lesiones Cardíacas/prevención & control , Histamina/análisis , Histidina Descarboxilasa/deficiencia , Histidina Descarboxilasa/metabolismo , Humanos , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Regiones Promotoras Genéticas , Pirilamina/farmacología
20.
Mol Cell ; 59(1): 50-61, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26028536

RESUMEN

MicroRNAs (miRNAs) are small non-coding RNAs, and they bind to complementary sequences in the three prime untranslated regions (3' UTRs) of target mRNA transcripts, thereby inhibiting mRNA translation or promoting mRNA degradation. Excessive reactive oxygen species (ROS) can cause cell-damaging effects through oxidative modification of macromolecules leading to their inappropriate functions. Such oxidative modification is related to cancers, aging, and neurodegenerative and cardiovascular diseases. Here we report that miRNAs can be oxidatively modified by ROS. We identified that miR-184 upon oxidative modification associates with the 3' UTRs of Bcl-xL and Bcl-w that are not its native targets. The mismatch of oxidized miR-184 with Bcl-xL and Bcl-w is involved in the initiation of apoptosis in the study with rat heart cell line H9c2 and mouse models. Our results reveal a model of ROS in regulating cellular events by oxidatively modifying miRNA.


Asunto(s)
Regiones no Traducidas 3'/genética , MicroARNs/metabolismo , Proteínas/genética , Especies Reactivas de Oxígeno/metabolismo , Proteína bcl-X/genética , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Línea Celular , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Miocardio/citología , Miocardio/metabolismo , Oxidación-Reducción , Interferencia de ARN , ARN Interferente Pequeño , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...