Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Microb Pathog ; 161(Pt B): 105284, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34767930

RESUMEN

Chlamydia pneumoniae (Cpn) has been reported to be involved in the pathogenesis of early atherosclerosis by inducing macrophage-derived foam cell formation in the presence of low-density lipoprotein (LDL). However, the biochemical mechanisms underlying Cpn-induced foam cell formation are still not fully elucidated. The present study showed that in LDL-treated THP-1-derived macrophages, Cpn not only upregulated the expression of scavenger receptor A1 (SR-A1) and acyl-coenzyme A: cholesterol acyltransferase 1 (ACAT1), but it also downregulated the expression of ATP binding cassette transporters (ABCA1 and ABCG1) at both the mRNA and protein levels. These processes facilitated cholesterol accumulation and promoted macrophage-derived foam cell formation. Treatment with the peroxisome proliferator-activated receptor (PPAR)-γ agonist rosiglitazone or the PPARα agonist fenofibrate decreased the number of foam cells induced by Cpn, while the PPARγ antagonist GW9662, the PPARα antagonist MK886, or PPARα/γ siRNAs enhanced the effect of Cpn on foam cell formation and gene expression of SR-A1, ACAT1, and ABCA1/G1. Moreover, the PPARγ agonist rosiglitazone reversed the downregulation of CD36 by Cpn, while PPARγ siRNA and the PPARγ inhibitor GW9662 further suppressed CD36 expression. However, the PPARα agonist, inhibitor, and siRNA all showed no effect on CD36 expression. In conclusion, the PPARα and PPARγ pathways are both involved in Cpn-induced macrophage-derived foam cell formation by upregulating SR-A1 and ACAT1 and downregulating ABCA1/G1 expression.


Asunto(s)
Chlamydophila pneumoniae , Células Espumosas , Transducción de Señal , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Aciltransferasas , Antígenos CD36/genética , Antígenos CD36/metabolismo , Chlamydophila pneumoniae/metabolismo , Regulación hacia Abajo , Células Espumosas/metabolismo , Células Espumosas/microbiología , Humanos , PPAR alfa/genética , PPAR gamma , Células THP-1 , Regulación hacia Arriba
2.
Int J Mol Sci ; 22(7)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33806236

RESUMEN

Classic atherosclerosis risk factors do not explain all cases of chronic heart disease. There is significant evidence that gut microbiota may influence the development of atherosclerosis. The widespread prevalence of chronic Helicobacter pylori (H. pylori, HP) infections suggests that HP can be the source of components that stimulate local and systemic inflammatory responses. Elevated production of reactive oxygen species during HP infection leads to cholesterol oxidation, which drives atherogenesis. The aim of this study is to explore the link between persistent HP infection and a high-fat diet in the development of proinflammatory conditions that are potentially proatherogenic. An in vivo model of Caviae porcellus infected with HP and exposed to an experimental diet was investigated for the occurrence of a proinflammatory and proatherogenic endothelial environment. Vascular endothelial primary cells exposed to HP components were tested in vitro for oxidative stress, cell activation and apoptosis. The infiltration of inflammatory cells into the vascular endothelium of animals infected with HP and exposed to a high-fat diet was observed in conjunction with an increased level of inflammatory markers systemically. The arteries of such animals were the least elastic, suggesting the role of HP in arterial stiffness. Soluble HP components induced transformation of macrophages to foam cells in vitro and influenced the endothelial life span, which was correlated with Collagen I upregulation. These preliminary results support the hypothesis that HP antigens act synergistically with a high-fat diet in the development of proatherogenic conditions.


Asunto(s)
Dieta Aterogénica , Dieta Alta en Grasa , Endotelio Vascular/metabolismo , Infecciones por Helicobacter/complicaciones , Animales , Anticuerpos Antibacterianos/inmunología , Aterosclerosis/etiología , Aterosclerosis/microbiología , Modelos Animales de Enfermedad , Células Endoteliales/microbiología , Femenino , Células Espumosas/metabolismo , Células Espumosas/microbiología , Gastritis/metabolismo , Gastritis/microbiología , Cobayas , Helicobacter pylori , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunoglobulina G , Inflamación , Macrófagos/metabolismo , Macrófagos/microbiología , Masculino , Rigidez Vascular
3.
PLoS Pathog ; 16(10): e1008929, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33002063

RESUMEN

The ability of Mycobacterium tuberculosis (Mtb) to persist inside host cells relies on metabolic adaptation, like the accumulation of lipid bodies (LBs) in the so-called foamy macrophages (FM), which are favorable to Mtb. The activation state of macrophages is tightly associated to different metabolic pathways, such as lipid metabolism, but whether differentiation towards FM differs between the macrophage activation profiles remains unclear. Here, we aimed to elucidate whether distinct macrophage activation states exposed to a tuberculosis-associated microenvironment or directly infected with Mtb can form FM. We showed that the triggering of signal transducer and activator of transcription 6 (STAT6) in interleukin (IL)-4-activated human macrophages (M(IL-4)) prevents FM formation induced by pleural effusion from patients with tuberculosis. In these cells, LBs are disrupted by lipolysis, and the released fatty acids enter the ß-oxidation (FAO) pathway fueling the generation of ATP in mitochondria. Accordingly, murine alveolar macrophages, which exhibit a predominant FAO metabolism, are less prone to become FM than bone marrow derived-macrophages. Interestingly, direct infection of M(IL-4) macrophages with Mtb results in the establishment of aerobic glycolytic pathway and FM formation, which could be prevented by FAO activation or inhibition of the hypoxia-inducible factor 1-alpha (HIF-1α)-induced glycolytic pathway. In conclusion, our results demonstrate that Mtb has a remarkable capacity to induce FM formation through the rewiring of metabolic pathways in human macrophages, including the STAT6-driven alternatively activated program. This study provides key insights into macrophage metabolism and pathogen subversion strategies.


Asunto(s)
Células Espumosas/microbiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metabolismo de los Lípidos , Macrófagos/microbiología , Mycobacterium tuberculosis/patogenicidad , Animales , Gotas Lipídicas/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/fisiología , Tuberculosis/microbiología
4.
PLoS Negl Trop Dis ; 14(10): e0008850, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33075048

RESUMEN

Leprosy is a chronic infectious disease caused by Mycobacterium leprae (M. leprae). In lepromatous leprosy (LL), skin macrophages, harboring extensive bacterial multiplication, gain a distinctive foamy appearance due to increased intracellular lipid load. To determine the mechanism by which M. leprae modifies the lipid homeostasis in host cells, an in vitro M. leprae infection system, using human macrophage precursor THP-1 cells and M. leprae prepared from the footpads of nude mice, was employed. RNA extracted from skin smear samples of patients was used to investigate host gene expressions before and after multidrug therapy (MDT). We found that a cluster of peroxisome proliferator-activated receptor (PPAR) target genes associated with adipocyte differentiation were strongly induced in M. leprae-infected THP-1 cells, with increased intracellular lipid accumulation. PPAR-δ and PPAR-γ expressions were induced by M. leprae infection in a bacterial load-dependent manner, and their proteins underwent nuclear translocalization after infection, indicating activation of PPAR signaling in host cells. Either PPAR-δ or PPAR-γ antagonist abolished the effect of M. leprae to modify host gene expressions and inhibited intracellular lipid accumulation in host cells. M. leprae-specific gene expressions were detected in the skin smear samples both before and after MDT, whereas PPAR target gene expressions were dramatically diminished after MDT. These results suggest that M. leprae infection activates host PPAR signaling to induce an array of adipocyte differentiation-associated genes, leading to accumulation of intracellular lipids to accommodate M. leprae parasitization. Certain PPAR target genes in skin lesions may serve as biomarkers for monitoring treatment efficacy.


Asunto(s)
Células Espumosas/microbiología , Lepra/metabolismo , Macrófagos/microbiología , Mycobacterium leprae/fisiología , PPAR delta/metabolismo , PPAR gamma/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Adipocitos/microbiología , Animales , Diferenciación Celular , Células Espumosas/metabolismo , Humanos , Leprostáticos/uso terapéutico , Lepra/tratamiento farmacológico , Lepra/genética , Lepra/microbiología , Metabolismo de los Lípidos , Macrófagos/metabolismo , Ratones , Ratones Desnudos , Mycobacterium leprae/efectos de los fármacos , PPAR delta/genética , PPAR gamma/genética , Piel/metabolismo , Piel/microbiología
5.
Front Immunol ; 11: 910, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32477367

RESUMEN

Tuberculosis (TB) is a leading cause of death worldwide following infection with Mycobacterium tuberculosis (Mtb), with 1.5 million deaths from this disease reported in 2018. Once the bacilli are inhaled, alveolar and interstitial macrophages become infected with Mtb and differentiate into lipid-laden foamy macrophages leading to lung inflammation. Thus, the presence of lipid-laden foamy macrophages is the hallmark of TB granuloma; these Mtb-infected foamy macrophages are the major niche for Mtb survival. The fate of TB pathogenesis is likely determined by the altered function of Mtb-infected macrophages, which initiate and mediate TB-related lung inflammation. As Mtb-infected foamy macrophages play central roles in the pathogenesis of Mtb, they may be important in the development of host-directed therapy against TB. Here, we summarize and discuss the current understanding of the alterations in alveolar and interstitial macrophages in the regulation of Mtb infection-induced immune responses. Metabolic reprogramming of lipid-laden foamy macrophages following Mtb infection or virulence factors are also summarized. Furthermore, we review the therapeutic interventions targeting immune responses and metabolic pathways, from in vitro, in vivo, and clinical studies. This review will further our understanding of the Mtb-infected foamy macrophages, which are both the major Mtb niche and therapeutic targets against TB.


Asunto(s)
Células Espumosas/microbiología , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/terapia , Animales , Reprogramación Celular , Interacciones Huésped-Patógeno , Humanos , Inflamación , Macrófagos Alveolares/microbiología , Ratones , Tuberculosis/microbiología
6.
Scand J Immunol ; 91(4): e12866, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31960452

RESUMEN

Understanding mechanisms of cavitation in tuberculosis (TB) is the missing link that could advance the field towards better control of the infection. Descriptions of human TB suggest that postprimary TB begins as lipid pneumonia of foamy macrophages that undergoes caseating necrosis and fragmentation to produce cavities. This study aimed to investigate the various mycobacterial antigens accumulating in foamy macrophages and their relation to tissue destruction and necrosis. Pulmonary tissues from mice with slowly progressive TB were studied for histopathology, acid-fast bacilli (AFB) and presence of mycobacterial antigens. Digital quantification using Aperio ImageScope was done. Until week 12 postinfection, mice were healthy, and lesions were small with scarce AFB and mycobacterial antigens. Colony-forming units (CFUs) increased exponentially. At week 16-33, mice were sick, macrophages attained foamy appearance with an increase in antigens (P < .05), 1.5 log increase in CFUs and an approximately onefold increase in AFB. At week 37-41, mice started dying with a shift in morphology towards necrosis. A >20-fold increase in mycobacterial antigens was observed with only less than one log increase in CFUs and sevenfold increase in AFB. Secreted antigens were significantly (P < .05) higher compared to cell-wall antigens throughout infection. Focal areas of necrosis were associated with an approximately 40-fold increase in antigen MPT46, functionally active thioredoxin, and a significant increase in all secreted antigens. In conclusion, mycobacterial antigens accumulate in the foamy macrophages in TB lesions during slowly progressive murine pulmonary TB. Secreted antigens and MPT46 correlated with necrosis, thereby implying that they might trigger the formation of cavities.


Asunto(s)
Antígenos Bacterianos/inmunología , Células Espumosas/inmunología , Células Espumosas/microbiología , Tuberculosis Pulmonar/patología , Animales , Células Espumosas/patología , Ratones , Mycobacterium tuberculosis , Necrosis , Tuberculosis Pulmonar/inmunología
7.
Biomedica ; 39(Supl. 2): 26-31, 2019 08 01.
Artículo en Inglés, Español | MEDLINE | ID: mdl-31529831

RESUMEN

Patients with lepromatous leprosy that have received treatment for many years usually get follow up biopsies for persistent skin lesions or positive bacilloscopy even if the values are lower than in the initial bacilloscopy. We report the case of a 48-year old woman with long-standing lepromatous leprosy of 15 years of evolution, with a bacterial index of 4 in the direct smear and the initial skin biopsy. The patient was treated with multidrug therapy for 32 months although the treatment recommended by the World Health Organization (WHO) is only for 12 months. A skin biopsy was taken to determine if there was an active disease. We observed a diffuse dermal inflammation with numerous foreign body giant cells and vacuolated macrophages (Virchow´s cells). These cells contained granular acid-fast material that was also positive with immunohistochemistry for BCG. There were fragmented bacilli and the BI was 2. These cells were also strongly positive for CD68. The biopsy was interpreted as a residual form of lepromatous leprosy that did not require further multidrug therapy. We have observed similar histological profiles in several cases. The lack of clinical data makes it a histological challenge. The accumulation of lipids in these giant cells is due to bacillary destruction and fusion of vacuolated macrophages. We discuss here the role of bacillary and host lipids in the pathogenesis of lepromatous leprosy. We concluded that there was no need to extend the 12-month multidrug therapy recommended by WHO.


Los pacientes con lepra lepromatosa (LL) que han recibido tratamiento durante años, usualmente tienen seguimiento con biopsias de piel para lesiones persistentes o con baciloscopia positiva, con valores menores a los iniciales. Presentamos una mujer de 48 años con LL de 15 años de evolución, con índice bacilar (IB) 4 en el extendido directo y en la biopsia, que recibió terapia multidroga durante 32 meses, aunque el tratamiento recomendado por la Organización mundial de la salud (OMS) es de 12 meses. Se tomó una biopsia de piel para determinar si la enfermedad estaba activa. Se observó inflamación dérmica difusa con numerosas células gigantes tipo cuerpo extraño y macrófagos vacuolados (células de Virchow). Estas células, CD68 positivas, contenían material granular ácido-alcohol resistente, positivo con inmunohistoquímica para BCG. Se encontraron bacilos fragmentados y el IB fue de 2. Se interpretó como una forma residual de LL y que la paciente no requería MDT adicional. Este perfil histológico lo hemos observado en casos similares. Sin datos clínicos estas biopsias son un reto diagnóstico. La acumulación de lípidos en estas células gigantes se debe a la destrucción bacilar y a la fusión de macrófagos vacuolados. Revisamos el papel de los lípidos del bacilo y del huésped en la patogénesis de la LL. En estos casos no es necesario extender los 12 meses de MDT recomendados por la OMS. En el seguimiento de los pacientes se recomienda contar con los hallazgos clínicos, la baciloscopia, la biopsia anual de piel y los títulos IgM anti-glicolípido fenólico.


Asunto(s)
Células Espumosas/patología , Células Gigantes de Cuerpo Extraño/patología , Lepra Lepromatosa/patología , Piel/patología , Antígenos CD/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Biopsia , Pared Celular/química , Quimioterapia Combinada , Femenino , Células Espumosas/química , Células Espumosas/microbiología , Células Gigantes de Cuerpo Extraño/química , Células Gigantes de Cuerpo Extraño/microbiología , Interacciones Huésped-Patógeno , Humanos , Leprostáticos/uso terapéutico , Lepra Lepromatosa/tratamiento farmacológico , Lípidos/análisis , Persona de Mediana Edad , Mycobacterium leprae/química , Mycobacterium leprae/aislamiento & purificación , Piel/microbiología , Vacuolas
8.
Biomédica (Bogotá) ; 39(supl.2): 26-31, ago. 2019. graf
Artículo en Español | LILACS | ID: biblio-1038825

RESUMEN

Resumen Los pacientes con lepra lepromatosa que han recibido tratamiento durante años, usualmente requieren seguimiento con biopsias de piel para detectar lesiones persistentes o si la baciloscopia es positiva, incluso si los valores son menores que los iniciales. Se presenta el caso de una mujer de 48 años de edad con lepra lepromatosa de 15 años de evolución, índice bacilar de 4 en el extendido directo y en la biopsia, que recibió tratamiento con múltiples medicamentos durante 32 meses, aunque lo recomendado por la Organización Mundial de la Salud (OMS) es una duración de 12 meses. Se tomó una biopsia de piel para determinar si la enfermedad estaba activa. Se observó inflamación dérmica difusa con numerosas células gigantes de tipo cuerpo extraño y macrófagos vacuolados (células de Virchow). Estas células, CD68 positivas, contenían material granular ácido-alcohol resistente positivo con inmunohistoquímica para BCG. Se encontraron bacilos fragmentados y el índice bacilar fue de 2. Se interpretó como una forma residual de lepra lepromatosa y se concluyó que la paciente no requería prolongar el tratamiento con múltiples medicamentos. Este perfil histológico se ha observado en casos similares, pero sin datos clínicos estas biopsias representan un reto diagnóstico. La acumulación de lípidos en estas células gigantes se debe a la destrucción bacilar y a la fusión de macrófagos vacuolados. Se revisó el papel de los lípidos del bacilo y del huésped en la patogenia de la lepra lepromatosa. En estos casos, no es necesario extender los 12 meses de tratamiento con múltiples medicamentos recomendados por la OMS. En el seguimiento de los pacientes, se recomienda contar con los hallazgos clínicos, la baciloscopia, la biopsia anual de piel y los títulos IgM antiglucolípido fenólico.


Abstract Patients with lepromatous leprosy that have received treatment for many years usually get follow up biopsies for persistent skin lesions or positive bacilloscopy even if the values are lower than in the initial bacilloscopy. We report the case of a 48-year old woman with long-standing lepromatous leprosy of 15 years of evolution, with a bacterial index of 4 in the direct smear and the initial skin biopsy. The patient was treated with multidrug therapy for 32 months although the treatment recommended by the World Health Organization (WHO) is only for 12 months. A skin biopsy was taken to determine if there was an active disease. We observed a diffuse dermal inflammation with numerous foreign body giant cells and vacuolated macrophages (Virchow´s cells). These cells contained granular acid-fast material that was also positive with immunohistochemistry for BCG. There were fragmented bacilli and the BI was 2. These cells were also strongly positive for CD68. The biopsy was interpreted as a residual form of lepromatous leprosy that did not require further multidrug therapy. We have observed similar histological profiles in several cases. The lack of clinical data makes it a histological challenge. The accumulation of lipids in these giant cells is due to bacillary destruction and fusion of vacuolated macrophages. We discuss here the role of bacillary and host lipids in the pathogenesis of lepromatous leprosy. We concluded that there was no need to extend the 12-month multidrug therapy recommended by WHO. Clinical findings, bacilloscopy, annual skin biopsy, and anti-phenolic glycolipid-I IgM titers are recommended procedures for the follow-up of these patients.


Asunto(s)
Femenino , Humanos , Persona de Mediana Edad , Piel/patología , Lepra Lepromatosa/patología , Células Gigantes de Cuerpo Extraño/patología , Células Espumosas/patología , Piel/microbiología , Vacuolas , Biopsia , Antígenos de Diferenciación Mielomonocítica/análisis , Lepra Lepromatosa/tratamiento farmacológico , Antígenos CD/análisis , Células Gigantes de Cuerpo Extraño/microbiología , Células Gigantes de Cuerpo Extraño/química , Pared Celular/química , Quimioterapia Combinada , Interacciones Huésped-Patógeno , Células Espumosas/microbiología , Células Espumosas/química , Leprostáticos/uso terapéutico , Lípidos/análisis , Mycobacterium leprae/aislamiento & purificación , Mycobacterium leprae/química
9.
J Mol Cell Cardiol ; 135: 40-51, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31352044

RESUMEN

BACKGROUND: Seroepidemiological studies have highlighted a positive relation between CagA-positive Helicobacter pylori (H. pylori), atherosclerosis and related clinic events. However, this link has not been well validated. The present study was designed to explore the role of H. pylori PMSS1 (a CagA-positive strain that can translocate CagA into host cells) and exosomal CagA in the progression of atherosclerosis. METHODS: To evaluate whether H. pylori accelerates or even induces atherosclerosis, H. pylori-infected C57/BL6 mice and ApoE-/- mice were maintained under different dietary conditions. To identify the role of H. pylori-infected gastric epithelial cells-derived exosomes (Hp-GES-EVs) and exosomal CagA in atherosclerosis, ApoE-/- mice were given intravenous or intraperitoneal injections of saline, GES-EVs, Hp-GES-EVs, and recombinant CagA protein (rCagA). FINDINGS: CagA-positive H. pylori PMSS1 infection does not induce but promotes macrophage-derived foam cell formation and augments atherosclerotic plaque growth and instability in two animal models. Meanwhile, circulating Hp-GES-EVs are taken up in aortic plaque, and CagA is secreted in Hp-GES-EVs. Furthermore, the CagA-containing EVs and rCagA exacerbates macrophage-derived foam cell formation and lesion development in vitro and in vivo, recapitulating the pro-atherogenic effects of CagA-positive H. pylori. Mechanistically, CagA suppresses the transcription of cholesterol efflux transporters by downregulating the expression of transcriptional factors PPARγ and LXRα and thus enhances foam cell formation. INTERPRETATION: These results may provide new insights into the role of exosomal CagA in the pathogenesis of CagA-positive H. pylori infection-related atherosclerosis. It is suggested that preventing and eradicating CagA-positive H. pylori infection could reduce the incidence of atherosclerosis and related events.


Asunto(s)
Antígenos Bacterianos/metabolismo , Aterosclerosis/metabolismo , Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Células Espumosas/metabolismo , Mucosa Gástrica/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Animales , Aterosclerosis/microbiología , Aterosclerosis/patología , Células Epiteliales/microbiología , Células Epiteliales/patología , Células Espumosas/microbiología , Células Espumosas/patología , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Ratones
10.
Front Immunol ; 10: 2874, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31969876

RESUMEN

Mycobacterium avium spp. paratuberculosis (MAP) is the causative agent of Johne's disease (JD), also known as paratuberculosis, in ruminants. The mechanisms of JD pathogenesis are not fully understood, but it is known that MAP subverts the host immune system by using macrophages as its primary reservoir. MAP infection in macrophages is often studied in healthy cows or experimentally infected calves, but reports on macrophages from naturally infected cows are lacking. In our study, primary monocyte-derived macrophages (MDMs) from cows diagnosed as positive (+) or negative (-) for JD were challenged in vitro with live MAP. Analysis using next-generation RNA sequencing revealed that macrophages from JD(+) cows did not present a definite pattern of response to MAP infection. Interestingly, a considerable number of genes, up to 1436, were differentially expressed in JD(-) macrophages. The signatures of the infection time course of 1, 4, 8, and 24 h revealed differential expression of ARG2, COL1A1, CCL2, CSF3, IL1A, IL6, IL10, PTGS2, PTX3, SOCS3, TNF, and TNFAIP6 among other genes, with major effects on host signaling pathways. While several immune pathways were affected by MAP, other pathways related to hepatic fibrosis/hepatic stellate cell activation, lipid homeostasis, such as LXR/RXR (liver X receptor/retinoid X receptor) activation pathways, and autoimmune diseases (rheumatoid arthritis or atherosclerosis) also responded to the presence of live MAP. Comparison of the profiles of the unchallenged MDMs from JD(+) vs. JD(-) cows showed that 868 genes were differentially expressed, suggesting that these genes were already affected before monocytes differentiated into macrophages. The downregulated genes predominantly modified the general cell metabolism by downregulating amino acid synthesis and affecting cholesterol biosynthesis and other energy production pathways while introducing a pro-fibrotic pattern associated with foam cells. The upregulated genes indicated that lipid homeostasis was already supporting fat storage in uninfected JD(+) MDMs. For JD(+) MDMs, differential gene expression expounds long-term mechanisms established during disease progression of paratuberculosis. Therefore, MAP could further promote disease persistence by influencing long-term macrophage behavior by using both tolerance and fat-storage states. This report contributes to a better understanding of MAP's controls over the immune cell response and mechanisms of MAP survival.


Asunto(s)
Células Espumosas/inmunología , Inmunidad Innata/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Mycobacterium avium subsp. paratuberculosis/inmunología , Paratuberculosis/inmunología , Transcriptoma/inmunología , Animales , Bovinos , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/microbiología , Células Espumosas/microbiología , Perfilación de la Expresión Génica/métodos , Tolerancia Inmunológica/inmunología , Monocitos/inmunología , Monocitos/microbiología , Fenotipo
11.
J Biol Chem ; 292(27): 11326-11335, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28515317

RESUMEN

Mycobacterium tuberculosis causes tuberculosis in humans and predominantly infects alveolar macrophages. To survive inside host lesions and to evade immune surveillance, this pathogen has developed many strategies. For example, M. tuberculosis uses host-derived lipids/fatty acids as nutrients for prolonged persistence within hypoxic host microenvironments. M. tuberculosis imports these metabolites through its respective transporters, and in the case of host fatty acids, a pertinent question arises: does M. tuberculosis have the enzyme(s) for cleavage of fatty acids from host lipids? We show herein that a previously uncharacterized membrane-associated M. tuberculosis protein encoded by Rv2672 is conserved exclusively in actinomycetes, exhibits both lipase and protease activities, is secreted into macrophages, and catalyzes host lipid hydrolysis. In light of these functions, we annotated Rv2672 as mycobacterial secreted hydrolase 1 (Msh1). Furthermore, we found that this enzyme is up-regulated both in an in vitro model of hypoxic stress and in a mouse model of M. tuberculosis infection, suggesting that the pathogen requires Msh1 under hypoxic conditions. Silencing Msh1 expression compromised the ability of M. tuberculosis to proliferate inside lipid-rich foamy macrophages but not under regular culture conditions in vitro, underscoring Msh1's importance for M. tuberculosis persistence in lipid-rich microenvironments. Of note, this is the first report providing insight into the mechanism of host lipid catabolism by an M. tuberculosis enzyme, augmenting our current understanding of how M. tuberculosis meets its nutrient requirements under hypoxic conditions.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Espumosas/metabolismo , Células Espumosas/microbiología , Hidrolasas/metabolismo , Mycobacterium tuberculosis/enzimología , Tuberculosis/enzimología , Animales , Hipoxia de la Célula , Células Espumosas/patología , Metabolismo de los Lípidos , Ratones , Mycobacterium tuberculosis/patogenicidad , Células RAW 264.7 , Tuberculosis/genética , Tuberculosis/patología
12.
Pathog Dis ; 74(6)2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27481727

RESUMEN

Macrophage foam cell formation is a key event in atherosclerosis. Several triggers induce low-density lipoprotein (LDL) uptake by macrophages to create foam cells, including infections with Porphyromonas gingivalis and Chlamydia pneumoniae, two pathogens that have been linked to atherosclerosis. While gene regulation during foam cell formation has been examined, comparative investigations to identify shared and specific pathogen-elicited molecular events relevant to foam cell formation are not well documented. We infected mouse bone marrow-derived macrophages with P. gingivalis or C. pneumoniae in the presence of LDL to induce foam cell formation, and examined gene expression using an atherosclerosis pathway targeted plate array. We found over 30 genes were significantly induced in response to both pathogens, including PPAR family members that are broadly important in atherosclerosis and matrix remodeling genes that may play a role in plaque development and stability. Six genes mainly involved in lipid transport were significantly downregulated. The response overall was remarkably similar and few genes were regulated in a pathogen-specific manner. Despite very divergent lifestyles, P. gingivalis and C. pneumoniae activate similar gene expression profiles during foam cell formation that may ultimately serve as targets for modulating infection-elicited foam cell burden, and progression of atherosclerosis.


Asunto(s)
Células Espumosas/metabolismo , Células Espumosas/patología , Interacciones Huésped-Patógeno , Macrófagos/metabolismo , Macrófagos/patología , Transducción de Señal , Animales , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Chlamydophila pneumoniae/inmunología , Análisis por Conglomerados , Biología Computacional/métodos , Citocinas/metabolismo , Células Espumosas/inmunología , Células Espumosas/microbiología , Perfilación de la Expresión Génica , Ontología de Genes , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Mediadores de Inflamación/metabolismo , Metabolismo de los Lípidos , Peroxidación de Lípido , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Anotación de Secuencia Molecular , Porphyromonas gingivalis/inmunología
13.
Cell Signal ; 28(9): 1292-1303, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27234131

RESUMEN

CD36, a scavenger receptor, plays an important role in the progression of atherosclerosis through its interaction with oxidized low-density lipoprotein (ox-LDL). Porphyromonas gingivalis (P. gingivalis, Pg) has been shown to promote macrophage-derived foam cell formation by affecting the expression of CD36. However, the regulatory role of CD36 in macrophages infected with Pg remains largely unknown. Therefore, the aim of the present study was to explore the molecular mechanism of Pg induced CD36 expression in macrophages. Our results showed that Pg promoted ox-LDL uptake by macrophages and the formation of foam cells. Pg infection increased CD36 mRNA and protein levels in ox-LDL-untreated macrophages. Moreover, small interferon RNA (siRNA) targeting CD36 significantly reduced foam cell formation induced by Pg. Additionally, Pg stimulated nuclear translocation of p65, which directly bound to the promoters of CD36 to facilitate its transcription. Inhibition of p65, NF-κB or ERK1/2 blocked Pg-induced CD36 production; whereas, overexpression of NF-κB subunits p65 and p50 upregulated CD36. Furthermore, Ras inhibitors significantly attenuated ERK1/2 activation and CD36 expression. Taken together, the data indicated that stimulation of the ERK/NF-κB pathway by Pg led to transactivation of the CD36 promoters, thereby upregulating CD36 expression in the infected macrophages. These findings may help design new treatment strategies in atherosclerosis.


Asunto(s)
Infecciones por Bacteroidaceae/genética , Infecciones por Bacteroidaceae/microbiología , Antígenos CD36/genética , Sistema de Señalización de MAP Quinasas , Macrófagos Peritoneales/microbiología , FN-kappa B/metabolismo , Porphyromonas gingivalis/fisiología , Regulación hacia Arriba/genética , Animales , Infecciones por Bacteroidaceae/patología , Antígenos CD36/metabolismo , Femenino , Células Espumosas/metabolismo , Células Espumosas/microbiología , Células Espumosas/patología , Células HEK293 , Humanos , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , Modelos Biológicos , Regiones Promotoras Genéticas/genética , Unión Proteica , Subunidades de Proteína/metabolismo , Células RAW 264.7
14.
Curr Opin Microbiol ; 29: 30-6, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26544033

RESUMEN

The interplay between Mycobacterium tuberculosis lipid metabolism, the immune response and lipid homeostasis in the host creates a complex and dynamic pathogen-host interaction. Advances in imaging and metabolic analysis techniques indicate that M. tuberculosis preferentially associates with foamy cells and employs multiple physiological systems to utilize exogenously derived fatty-acids and cholesterol. Moreover, novel insights into specific host pathways that control lipid accumulation during infection, such as the PPARγ and LXR transcriptional regulators, have begun to reveal mechanisms by which host immunity alters the bacterial micro-environment. As bacterial lipid metabolism and host lipid regulatory pathways are both important, yet inherently complex, components of active tuberculosis, delineating the heterogeneity in lipid trafficking within disease states remains a major challenge for therapeutic design.


Asunto(s)
Interacciones Huésped-Patógeno , Metabolismo de los Lípidos , Macrófagos/microbiología , Mycobacterium tuberculosis/metabolismo , Tuberculosis/metabolismo , Tuberculosis/microbiología , Animales , Ácidos Grasos/metabolismo , Células Espumosas/metabolismo , Células Espumosas/microbiología , Homeostasis , Humanos , Gotas Lipídicas/metabolismo , Macrófagos/metabolismo , Macrófagos/ultraestructura , Redes y Vías Metabólicas/fisiología , Ratones , Mycobacterium tuberculosis/crecimiento & desarrollo , Tuberculosis/terapia
15.
Atherosclerosis ; 235(2): 519-25, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24953492

RESUMEN

OBJECTIVES: ATP-binding cassette transporter A1 (ABCA1) is critical in exporting cholesterol from macrophages and plays a protective role in the development of atherosclerosis. This study was to determine the effects and potential mechanisms of Chlamydia pneumoniae (C. pneumoniae) on ABCA1 expression and cellular cholesterol efflux in THP-1 macrophage-derived foam cells. METHODS AND RESULTS: C. pneumoniae significantly decreased the expression of ABCA1 and reduced cholesterol efflux. Furthermore, we found that C. pneumoniae suppressed ABCA1 expression via up-regulation of miR-33s. The inhibition of C. pneumoniae-induced NF-κB activation decreased miR-33s expression and enhanced ABCA1 expression. In addition, C. pneumoniae increased Toll-like receptor 2 (TLR2) expressions, inhibition of which by siRNA could also block NF-κB activation and miR-33s expression, and promot the expression of ABCA1. CONCLUSION: Taken together, these results reveal that C. pneumoniae may negatively regulate ABCA1 expression via TLR2-NF-κB and miR-33 pathways in THP-1 macrophage-derived foam cells, which may provide new insights for understanding the effects of C. pneumoniae on the pathogenesis of atherosclerosis.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/biosíntesis , Chlamydophila pneumoniae/fisiología , Células Espumosas/metabolismo , MicroARNs/fisiología , FN-kappa B/fisiología , Receptor Toll-Like 2/fisiología , Colesterol/metabolismo , Células Espumosas/microbiología , Humanos , Macrófagos/metabolismo
16.
Microb Pathog ; 69-70: 1-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24657322

RESUMEN

Chlamydia pneumoniae (C. pneumoniae) is now widely accepted as an independent risk of atherosclerosis development. In this paper, our results showed that C. pneumoniae infection significantly increased the number of foam cells in LDL-treated THP-1 macrophages. C-Jun NH2 terminal kinase (JNK1/2) inhibitor SP600125 and extracellular signal-regulated kinase (ERK1/2) inhibitor PD98059 strongly inhibited C. pneumoniae-induced accumulation of lipid droplet, whereas p38 inhibitor SB203580 had no obvious effect on lipid accumulation. Furthermore, we found that C. pneumoniae not only stimulated the phosphorylation of Mitogen-activated protein kinase (MAPK) including JNK1/2, ERK1/2 and p38 but also down-regulated the expression of peroxisome proliferator-activated receptors (PPARγ and PPARα) at mRNA and protein levels. However, the phosphorylation of JNK1/2, ERK1/2 and p38 MAPK by C. pneumoniae was substantially reversed after PPARγ agonist (rosiglitazone) or PPARα agonist (fenofibrate) treatment while PPARγ inhibitor (GW9662) and PPARα antagonist (MK886) enhanced C. pneumoniae-induced phosphorylation of JNK1/2, ERK1/2 and p38. In addition, we demonstrated that C. pneumoniae-induced PPARγ and PPARα down-regulation were significantly suppressed by JNK1/2 inhibitor (SP600125) and ERK1/2 inhibitor (PD98059), but not p38 inhibitor (SB203580). These results first declare that MAPK-PPARα/γ reciprocal signal pathways are involved in C. pneumoniae, which induces foam cell formation, thus facilitating atherogenesis.


Asunto(s)
Chlamydophila pneumoniae/inmunología , Células Espumosas/inmunología , Células Espumosas/microbiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Transducción de Señal , Línea Celular , Humanos , Macrófagos/inmunología , Macrófagos/microbiología
18.
Mol Oral Microbiol ; 28(1): 28-39, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23194377

RESUMEN

Clinical studies and experimental modeling identify a potential link between periodontal disease and periodontal pathogens such as Porphyromonas gingivalis and atherosclerosis and formation of macrophage foam cells. Toll-like receptors and molecules governing their intracellular signaling pathways such as MyD88 play roles in atherosclerosis, as well as host response to P. gingivalis. The aim of this study was to define roles of MyD88 and TRIF during macrophage foam cell formation in response to P. gingivalis. In the presence of human low-density lipoprotein (LDL) mouse bone-marrow-derived macrophages (BMφ) cultured with P. gingivalis responded with significant reduction in tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). The BMφ stained strongly with oil red O, regardless of whether bacterial challenge occurred concurrent with or before LDL treatment. Heat-killed P. gingivalis stimulated foam cell formation in a similar way to live bacteria. The BMφ from MyD88-knockout and Lps2 mice revealed a significant role for MyD88, and a minor role for TRIF in P. gingivalis-elicited foam cell formation. Porphyromonas gingivalis-elicited TNF-α and IL-6 were affected by MyD88 ablation and to a lesser extent by TRIF status. These data indicate that LDL affects the TNF-α and IL-6 response of macrophages to P. gingivalis challenge and that MyD88 and TRIF play important roles in P. gingivalis-elicited foam cell formation.


Asunto(s)
Células Espumosas/microbiología , Macrófagos/microbiología , Factor 88 de Diferenciación Mieloide/inmunología , Porphyromonas gingivalis/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Compuestos Azo , Proteínas Bacterianas/genética , Células de la Médula Ósea , Células Cultivadas , Técnicas de Cocultivo , Colorantes , Células Espumosas/inmunología , Humanos , Mediadores de Inflamación/inmunología , Interleucina-6/inmunología , Lipoproteínas LDL/farmacología , Macrófagos/inmunología , Manosiltransferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Mutación Puntual/genética , Factor de Necrosis Tumoral alfa/inmunología
19.
Microbes Infect ; 14(13): 1227-37, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22982567

RESUMEN

Lipid inclusions play an important role in several pathological processes. Intracellular bacterial pathogens, such as members of the Mycobacterium and Chlamydia species are able to trigger the formation of lipid-laden foamy macrophages. Lipid droplet accumulation in the host constitutes a reservoir used by the bacilli for long-term persistence. Viruses need lipid droplets as assembly platform. We present the current knowledge about structural, functional and regulatory aspects of lipid inclusions.


Asunto(s)
Células Espumosas/microbiología , Cuerpos de Inclusión/metabolismo , Lípidos/química , Macrófagos/microbiología , Animales , Chlamydia/fisiología , Infecciones por Chlamydia/metabolismo , Infecciones por Chlamydia/microbiología , Citosol/metabolismo , Interacciones Huésped-Patógeno , Humanos , Metabolismo de los Lípidos , Lípidos/genética , Mycobacterium/fisiología , Infecciones por Mycobacterium/metabolismo , Infecciones por Mycobacterium/microbiología , Virosis/metabolismo , Virosis/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...