Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 19(8): e1011573, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37624851

RESUMEN

Pseudomonas aeruginosa (P.a.) infection accounts for nearly 20% of all cases of hospital acquired pneumonia with mortality rates >30%. P.a. infection induces a robust inflammatory response, which ideally enhances bacterial clearance. Unfortunately, excessive inflammation can also have negative effects, and often leads to cardiac dysfunction with associated morbidity and mortality. However, it remains unclear how P.a. lung infection causes cardiac dysfunction. Using a murine pneumonia model, we found that P.a. infection of the lungs led to severe cardiac left ventricular dysfunction and electrical abnormalities. More specifically, we found that neutrophil recruitment and release of S100A8/A9 in the lungs activates the TLR4/RAGE signaling pathways, which in turn enhance systemic inflammation and subsequent cardiac dysfunction. Paradoxically, global deletion of S100A8/A9 did not improve but aggravated cardiac dysfunction and mortality likely due to uncontrolled bacterial burden in the lungs and heart. Our results indicate that P.a. infection induced release of S100A8/9 is double-edged, providing increased risk for cardiac dysfunction yet limiting P.a. growth.


Asunto(s)
Cardiopatías , Infecciones por Pseudomonas , Animales , Ratones , Pseudomonas aeruginosa , Corazón , Inflamación , Pulmón
2.
Tuberculosis (Edinb) ; 142: 102377, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37531864

RESUMEN

The Many Hosts of Mycobacteria (MHM) meeting series brings together basic scientists, clinicians and veterinarians to promote robust discussion and dissemination of recent advances in our knowledge of numerous mycobacterial diseases, including human and bovine tuberculosis (TB), nontuberculous mycobacteria (NTM) infection, Hansen's disease (leprosy), Buruli ulcer and Johne's disease. The 9th MHM conference (MHM9) was held in July 2022 at The Ohio State University (OSU) and centered around the theme of "Confounders of Mycobacterial Disease." Confounders can and often do drive the transmission of mycobacterial diseases, as well as impact surveillance and treatment outcomes. Various confounders were presented and discussed at MHM9 including those that originate from the host (comorbidities and coinfections) as well as those arising from the environment (e.g., zoonotic exposures), economic inequality (e.g. healthcare disparities), stigma (a confounder of leprosy and TB for millennia), and historical neglect (a confounder in Native American Nations). This conference report summarizes select talks given at MHM9 highlighting recent research advances, as well as talks regarding the historic and ongoing impact of TB and other infectious diseases on Native American Nations, including those in Southwestern Alaska where the regional TB incidence rate is among the highest in the Western hemisphere.


Asunto(s)
Coinfección , Infecciones por Mycobacterium no Tuberculosas , Mycobacterium tuberculosis , Tuberculosis Bovina , Animales , Bovinos , Humanos , Micobacterias no Tuberculosas , Infecciones por Mycobacterium no Tuberculosas/microbiología
4.
iScience ; 25(12): 105701, 2022 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-36474635

RESUMEN

Influenza A virus (IAV) and SARS-CoV-2 virus are both acute respiratory viruses currently circulating in the human population. This study aims to determine the impact of IAV infection on SARS-CoV-2 pathogenesis and cardiomyocyte function. Infection of human bronchial epithelial cells (HBEC), A549 cells, lung fibroblasts (HLF), monocyte derived macrophages (MDMs), cardiac fibroblasts (HCF) and hiPSC-derived cardiomyocytes with IAV enhanced the expression of ACE2, the SARS-CoV-2 receptor. Similarly, IAV infection increased levels of ACE2 in the lungs of mice and humans. Of interest, we detected heavily glycosylated form of ACE2 in hiPSC-CMs and poorly glycosylated ACE2 in other cell types. Also, prior IAV infection enhances SARS-CoV-2 spike protein binding and viral entry in all cell types. However, efficient SARS-CoV-2 replication was uniquely inhibited in cardiomyocytes. Glycosylation of ACE2 correlated with enzymatic conversion of its substrate Ang II, induction of eNOS and nitric oxide production, may provide a potential mechanism for the restricted SARS-CoV-2 replication in cardiomyocytes.

5.
Front Cell Infect Microbiol ; 12: 990402, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36189368

RESUMEN

Age is a major risk factor for chronic infections, including tuberculosis (TB). Elderly TB patients also suffer from elevated levels of psychological stress. It is not clear how psychological stress impacts immune response to Mycobacterium tuberculosis (M.tb). In this study, we used social disruption stress (SDR) to investigate effects of psychological stress in young and old mice. Unexpectedly, we found that SDR suppresses lung inflammation in old mice as evidenced by lower pro-inflammatory cytokine levels in bronchial lavage fluid and decreased cytokine mRNA expression by alveolar macrophages. To investigate effects of stress on M.tb infection, mice were subjected to SDR and then infected with M.tb. As previously reported, old mice were better at controlling infection at 30 days than young mice. This control was transient as CFUs at 60 days were higher in old control mice compared to young mice. Consistently, SDR significantly increased M.tb growth at 60 days in old mice compared to young mice. In addition, SDR in old mice resulted in accumulation of IL-10 mRNA and decreased IFN-γ mRNA at 60 days. Also, confocal microscopy of lung sections from old SDR mice showed increased number of CD4 T cells which express LAG3 and CD49b, markers of IL-10 secreting regulatory T cells. Further, we also demonstrated that CD4 T cells from old SDR mice express IL-10. Thus, we conclude that psychological stress in old mice prior to infection, increases differentiation of IL-10 secreting T cells, which over time results in loss of control of the infection.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Citocinas/metabolismo , Integrina alfa2 , Interleucina-10/genética , Pulmón/metabolismo , Ratones , ARN Mensajero , Estrés Psicológico
6.
Sci Adv ; 8(19): eabm5371, 2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35544568

RESUMEN

Cardiac dysfunction is a common complication of severe influenza virus infection, but whether this occurs due to direct infection of cardiac tissue or indirectly through systemic lung inflammation remains unclear. To test the etiology of this aspect of influenza disease, we generated a novel recombinant heart-attenuated influenza virus via genome incorporation of target sequences for miRNAs expressed in cardiomyocytes. Compared with control virus, mice infected with miR-targeted virus had significantly reduced heart viral titers, confirming cardiac attenuation of viral replication. However, this virus was fully replicative in the lungs and induced similar systemic inflammation and weight loss compared to control virus. The miR-targeted virus induced fewer cardiac conduction irregularities and significantly less fibrosis in mice lacking interferon-induced transmembrane protein 3 (IFITM3), which serve as a model for influenza-associated cardiac pathology. We conclude that robust virus replication in the heart is required for pathology, even when lung inflammation is severe.


Asunto(s)
Gripe Humana , MicroARNs , Animales , Fibrosis , Humanos , Ratones , MicroARNs/genética , Miocitos Cardíacos , Replicación Viral/genética
7.
iScience ; 24(11): 103337, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34816100

RESUMEN

Lipopolysaccharides (LPSs) cause lethal endotoxemia if not rapidly cleared from blood circulation. Liver sinusoidal endothelial cells (LSEC) systemically clear LPS by unknown mechanisms. We discovered that LPS clearance through LSEC involves endocytosis and lysosomal inactivation via Stabilin-1 and 2 (Stab1 and Stab2) but does not involve TLR4. Cytokine production was inversely related to clearance/endocytosis of LPS by LSEC. When exposed to LPS, Stabilin double knockout mice (Stab DK) and Stab1 KO, but not Stab2 KO, showed significantly enhanced systemic inflammatory cytokine production and early death compared with WT mice. Stab1 KO is not significantly different from Stab DK in circulatory LPS clearance, LPS uptake and endocytosis by LSEC, and cytokine production. These data indicate that (1) Stab1 receptor primarily facilitates the proactive clearance of LPS and limits TLR4-mediated inflammation and (2) TLR4 and Stab1 are functionally opposing LPS receptors. These findings suggest that endotoxemia can be controlled by optimizing LPS clearance by Stab1.

8.
Aging Cell ; 20(8): e13438, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34342127

RESUMEN

Aging-mediated immune dysregulation affects the normal cardiac immune cell phenotypes and functions, resulting in cardiac distress. During cardiac inflammation, immune activation is critical for mounting the regenerative responses to maintain normal heart function. We investigated the impact of aging on myeloid cell phenotype and function during cardiac inflammation induced by a sub-lethal dose of LPS. Our data show that hearts of old mice contain more myeloid cells than the hearts of young mice. However, while the number of monocytic-derived suppressor cells did not differ between young and old mice, monocytic-derived suppressor cells from old mice were less able to suppress T-cell proliferation. Since cardiac resident macrophages (CRMs) are important for immune surveillance, clearance of dead cells, and tissue repair, we focused our studies on CRMs phenotype and function during steady state and LPS treatment. In the steady state, we observed significantly more MHC-IIlow and MHC-IIhigh CRMs in the hearts of old mice; however, these populations were decreased in both young and aged mice upon LPS treatment and the decrease in CRM populations correlated with defects in cardiac electrical activity. Notably, mice treated with a liver X receptor (LXR) agonist showed an increase in MerTK expression in CRMs of both young and old mice, which resulted in the reversal of cardiac electrical dysfunction caused by lipopolysaccharide (LPS). We conclude that aging alters the phenotype of CRMs, which contributes to the dysregulation of cardiac electrical dysfunction during infection in aged mice.


Asunto(s)
Envejecimiento/genética , Corazón/fisiopatología , Inflamación/fisiopatología , Macrófagos/metabolismo , Animales , Humanos , Ratones , Fenotipo
9.
J Immunol ; 206(6): 1284-1296, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33568400

RESUMEN

Neutralizing Abs suppress HIV infection by accelerating viral clearance from blood circulation in addition to neutralization. The elimination mechanism is largely unknown. We determined that human liver sinusoidal endothelial cells (LSEC) express FcγRIIb as the lone Fcγ receptor, and using humanized FcγRIIb mouse, we found that Ab-opsonized HIV pseudoviruses were cleared considerably faster from circulation than HIV by LSEC FcγRIIb. Compared with humanized FcγRIIb-expressing mice, HIV clearance was significantly slower in FcγRIIb knockout mice. Interestingly, a pentamix of neutralizing Abs cleared HIV faster compared with hyperimmune anti-HIV Ig (HIVIG), although the HIV Ab/Ag ratio was higher in immune complexes made of HIVIG and HIV than pentamix and HIV. The effector mechanism of LSEC FcγRIIb was identified to be endocytosis. Once endocytosed, both Ab-opsonized HIV pseudoviruses and HIV localized to lysosomes. This suggests that clearance of HIV, endocytosis, and lysosomal trafficking within LSEC occur sequentially and that the clearance rate may influence downstream events. Most importantly, we have identified LSEC FcγRIIb-mediated endocytosis to be the Fc effector mechanism to eliminate cell-free HIV by Abs, which could inform development of HIV vaccine and Ab therapy.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Endocitosis/inmunología , Células Endoteliales/inmunología , Infecciones por VIH/inmunología , Receptores de IgG/metabolismo , Animales , Capilares/citología , Capilares/inmunología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/virología , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Células HEK293 , VIH/inmunología , Infecciones por VIH/sangre , Infecciones por VIH/patología , Infecciones por VIH/virología , Voluntarios Sanos , Humanos , Hígado/irrigación sanguínea , Hígado/inmunología , Lisosomas/metabolismo , Lisosomas/virología , Masculino , Ratones , Ratones Noqueados , Cultivo Primario de Células , Receptores de IgG/genética
10.
J Exp Med ; 217(4)2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-31999304

RESUMEN

Aberrant NLRP3 inflammasome activation contributes to the development of endotoxemia. The importance of negative regulation of NLRP3 inflammasomes remains poorly understood. Here, we show that the E3 ubiquitin ligase Cbl-b is essential for preventing endotoxemia induced by a sub-lethal dose of LPS via a caspase-11/NLRP3-dependent manner. Further studies show that NLRP3 undergoes both K63- and K48-linked polyubiquitination. Cbl-b binds to the K63-ubiquitin chains attached to the NLRP3 leucine-rich repeat domain (LRR) via its ubiquitin-associated region (UBA) and then targets NLRP3 at K496 for K48-linked ubiquitination and proteasome-mediated degradation. We also identify RNF125 as an additional E3 ubiquitin ligase that initiates K63-linked ubiquitination of the NLRP3 LRR domain. Therefore, NLRP3 is sequentially ubiquitinated by K63- and K48-linked ubiquitination, thus keeping the NLRP3 inflammasomes in check and restraining endotoxemia.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endotoxemia/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo
11.
Cells ; 10(1)2020 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-33396359

RESUMEN

The immune system plays a pivotal role in the initiation, development and resolution of inflammation following insult or damage to organs. The heart is a vital organ which supplies nutrients and oxygen to all parts of the body. Heart failure (HF) has been conventionally described as a disease associated with cardiac tissue damage caused by systemic inflammation, arrhythmia and conduction defects. Cardiac inflammation and subsequent tissue damage is orchestrated by the infiltration and activation of various immune cells including neutrophils, monocytes, macrophages, eosinophils, mast cells, natural killer cells, and T and B cells into the myocardium. After tissue injury, monocytes and tissue-resident macrophages undergo marked phenotypic and functional changes, and function as key regulators of tissue repair, regeneration and fibrosis. Disturbance in resident macrophage functions such as uncontrolled production of inflammatory cytokines, growth factors and inefficient generation of an anti-inflammatory response or unsuccessful communication between macrophages and epithelial and endothelial cells and fibroblasts can lead to aberrant repair, persistent injury, and HF. Therefore, in this review, we discuss the role of cardiac macrophages on cardiac inflammation, tissue repair, regeneration and fibrosis.


Asunto(s)
Fibrosis/metabolismo , Lesiones Cardíacas/metabolismo , Macrófagos/metabolismo , Regeneración , Animales , Arritmias Cardíacas/inmunología , Arritmias Cardíacas/metabolismo , Citocinas/metabolismo , Cardiomiopatías Diabéticas/inmunología , Cardiomiopatías Diabéticas/metabolismo , Fibrosis/inmunología , Corazón/fisiopatología , Lesiones Cardíacas/inmunología , Homeostasis , Humanos , Hipertensión/inmunología , Hipertensión/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Macrófagos/citología , Mitocondrias/inmunología , Miocardio/citología , Miocardio/inmunología
12.
J Immunol ; 203(8): 2252-2264, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31511357

RESUMEN

The elderly population is more susceptible to pulmonary infections, including tuberculosis. In this article, we characterize the impact of aging on the phenotype of mouse alveolar macrophages (AMs) and their response to Mycobacterium tuberculosis. Uninfected AMs were isolated from bronchoalveolar lavage of young (3 mo) and old (18 mo) C57BL/6 mice. AMs from old mice expressed higher mRNA levels of CCL2, IFN-ß, IL-10, IL-12p40, TNF-α, and MIF than young mice, and old mice contained higher levels of CCL2, IL-1ß, IFN-ß, and MIF in their alveolar lining fluid. We identified two distinct AM subpopulations, a major CD11c+ CD11b- population and a minor CD11c+ CD11b+ population; the latter was significantly increased in old mice (4-fold). Expression of CD206, TLR2, CD16/CD32, MHC class II, and CD86 was higher in CD11c+ CD11b+ AMs, and these cells expressed monocytic markers Ly6C, CX3CR1, and CD115, suggesting monocytic origin. Sorted CD11c+ CD11b+ AMs from old mice expressed higher mRNA levels of CCL2, IL-1ß, and IL-6, whereas CD11c+ CD11b- AMs expressed higher mRNA levels of immune-regulatory cytokines IFN-ß and IL-10. CD11c+ CD11b+ AMs phagocytosed significantly more M. tuberculosis, which expressed higher RNA levels of genes required for M. tuberculosis survival. Our studies identify two distinct AM populations in old mice: a resident population and an increased CD11c+ CD11b+ AM subpopulation expressing monocytic markers, a unique inflammatory signature, and enhanced M. tuberculosis phagocytosis and survival when compared with resident CD11c+ CD11b- AMs, which are more immune regulatory in nature.


Asunto(s)
Inflamación/inmunología , Macrófagos Alveolares/inmunología , Tuberculosis/inmunología , Animales , Citocinas/inmunología , Femenino , Inflamación/patología , Macrófagos Alveolares/patología , Ratones , Ratones Endogámicos C57BL , Tuberculosis/patología
13.
Proc Natl Acad Sci U S A ; 116(37): 18607-18612, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31451661

RESUMEN

Influenza virus can disseminate from the lungs to the heart in severe infections and can induce cardiac pathology, but this has been difficult to study due to a lack of small animal models. In humans, polymorphisms in the gene encoding the antiviral restriction factor IFN-induced transmembrane protein 3 (IFITM3) are associated with susceptibility to severe influenza, but whether IFITM3 deficiencies contribute to cardiac dysfunction during infection is unclear. We show that IFITM3 deficiency in a new knockout (KO) mouse model increases weight loss and mortality following influenza virus infections. We investigated this enhanced pathogenesis with the A/PR/8/34 (H1N1) (PR8) influenza virus strain, which is lethal in KO mice even at low doses, and observed increased replication of virus in the lungs, spleens, and hearts of KO mice compared with wild-type (WT) mice. Infected IFITM3 KO mice developed aberrant cardiac electrical activity, including decreased heart rate and irregular, arrhythmic RR (interbeat) intervals, whereas WT mice exhibited a mild decrease in heart rate without irregular RR intervals. Cardiac electrical dysfunction in PR8-infected KO mice was accompanied by increased activation of fibrotic pathways and fibrotic lesions in the heart. Infection with a sublethal dose of a less virulent influenza virus strain (A/WSN/33 [H1N1]) resulted in a milder cardiac electrical dysfunction in KO mice that subsided as the mice recovered. Our findings reveal an essential role for IFITM3 in limiting influenza virus replication and pathogenesis in heart tissue and establish IFITM3 KO mice as a powerful model for studying mild and severe influenza virus-induced cardiac dysfunction.


Asunto(s)
Cardiopatías/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/genética , Proteínas de la Membrana/genética , Miocardio/patología , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Electrocardiografía , Fibrosis , Predisposición Genética a la Enfermedad , Corazón/diagnóstico por imagen , Corazón/virología , Cardiopatías/diagnóstico , Cardiopatías/patología , Cardiopatías/virología , Humanos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/complicaciones , Gripe Humana/inmunología , Gripe Humana/virología , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Índice de Severidad de la Enfermedad , Replicación Viral/genética , Replicación Viral/inmunología
14.
J Infect Dis ; 220(12): 1989-1998, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31412123

RESUMEN

BACKGROUND: Tuberculosis is caused by Mycobacterium tuberculosis. Recent emergence of multidrug-resistant (MDR) tuberculosis strains seriously threatens tuberculosis control and prevention. However, the role of macrophage multidrug resistance gene MDR1 on intracellular M. tuberculosis survival during antituberculosis drug treatment is not known. METHODS: We used the human monocyte-derived macrophages to study the role of M. tuberculosis in regulation of MDR1 and drug resistance. RESULTS: We discovered that M. tuberculosis infection increases the expression of macrophage MDR1 to extrude various chemical substances, including tuberculosis drugs, resulting in enhanced survival of intracellular M. tuberculosis. The pathway of regulation involves M. tuberculosis infection of macrophages and suppression of heat shock factor 1, a transcriptional regulator of MDR1 through the up-regulation of miR-431. Notably, nonpathogenic Mycobacterium smegmatis did not increase MDR1 expression, indicating active secretion of virulence factors in pathogenic M. tuberculosis contributing to this phenotype. Finally, inhibition of MDR1 improves antibiotic-mediated killing of M. tuberculosis. CONCLUSION: We report a novel finding that M. tuberculosis up-regulates MDR1 during infection, which limits the exposure of M. tuberculosis to sublethal concentrations of antimicrobials. This condition promotes M. tuberculosis survival and potentially enhances the emergence of resistant variants.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Regulación de la Expresión Génica , Macrófagos/metabolismo , Macrófagos/microbiología , Mycobacterium tuberculosis/fisiología , Tuberculosis/genética , Tuberculosis/microbiología , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Macrófagos/inmunología , Ratones , MicroARNs/genética , Viabilidad Microbiana/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Tuberculosis/metabolismo , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/metabolismo , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología , Factores de Virulencia
15.
Aging Cell ; 18(3): e12926, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30834643

RESUMEN

Biological aging dynamically alters normal immune and cardiac function, favoring the production of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α) and increased instances of cardiac distress. Cardiac failure is the primary reason for hospitalization of the elderly (65+ years). The elderly are also increasingly susceptible to developing chronic bacterial infections due to aging associated immune abnormalities. Since bacterial infections compound the rates of cardiac failure in the elderly, and this phenomenon is not entirely understood, the interplay between the immune system and cardiovascular function in the elderly is of great interest. Using Mycobacterium avium, an opportunistic pathogen, we investigated the effect of mycobacteria on cardiac function in aged mice. Young (2-3 months) and old (18-20 months) C57BL/6 mice were intranasally infected with M. avium strain 104, and we compared the bacterial burden, immune status, cardiac electrical activity, pathology, and function of infected mice against uninfected age-matched controls. Herein, we show that biological aging may predispose old mice infected with M. avium to mycobacterial dissemination into the heart tissue and this leads to cardiac dysfunction. M. avium infected old mice had significant dysrhythmia, cardiac hypertrophy, increased recruitment of CD45+ leukocytes, cardiac fibrosis, and increased expression of inflammatory genes in isolated heart tissue. This is the first study to report the effect of mycobacteria on cardiac function in an aged model. Our findings are critical to understanding how nontuberculous mycobacterium (NTM) and other mycobacterial infections contribute to cardiac dysfunction in the elderly population.


Asunto(s)
Arritmias Cardíacas/microbiología , Cardiomegalia/microbiología , Fibrosis Endomiocárdica/microbiología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Micobacterias no Tuberculosas , Envejecimiento/inmunología , Envejecimiento/patología , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Cardiomegalia/genética , Cardiomegalia/metabolismo , Susceptibilidad a Enfermedades , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Inflamación/microbiología , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Antígenos Comunes de Leucocito/inmunología , Ratones , Ratones Endogámicos C57BL , Infecciones por Mycobacterium no Tuberculosas/patología , Mycobacterium avium , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
16.
J Infect Dis ; 220(3): 514-523, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-30923818

RESUMEN

As we age, there is an increased risk for the development of tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) infection. Few studies consider that age-associated changes in the alveolar lining fluid (ALF) may increase susceptibility by altering soluble mediators of innate immunity. We assessed the impact of adult or elderly human ALF during Mtb infection in vitro and in vivo. We identified amplification of pro-oxidative and proinflammatory pathways in elderly ALF and decreased binding capability of surfactant-associated surfactant protein A (SP-A) and surfactant protein D (SP-D) to Mtb. Human macrophages infected with elderly ALF-exposed Mtb had reduced control and fewer phagosome-lysosome fusion events, which was reversed when elderly ALF was replenished with functional SP-A/SP-D. In vivo, exposure to elderly ALF exacerbated Mtb infection in young mice. Our studies demonstrate how the pulmonary environment changes as we age and suggest that Mtb may benefit from declining host defenses in the lung mucosa of the elderly.


Asunto(s)
Pulmón/inmunología , Pulmón/microbiología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Tuberculosis/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Inmunidad Innata/inmunología , Inflamación/inmunología , Inflamación/microbiología , Lisosomas/inmunología , Lisosomas/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Persona de Mediana Edad , Mycobacterium tuberculosis/inmunología , Fagosomas/inmunología , Fagosomas/microbiología , Proteína A Asociada a Surfactante Pulmonar/inmunología , Proteína D Asociada a Surfactante Pulmonar/inmunología , Tuberculosis/microbiología , Adulto Joven
17.
JACC Basic Transl Sci ; 3(5): 675-689, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30456339

RESUMEN

Ankyrin polypeptides are intracellular proteins responsible for targeting cardiac membrane proteins. Here, the authors demonstrate that ankyrin-G plays an unexpected role in normal compensatory physiological remodeling in response to myocardial stress and aging; the authors implicate disruption of ankyrin-G in human heart failure. Mechanistically, the authors illustrate that ankyrin-G serves as a key nodal protein required for cardiac myofilament integration with the intercalated disc. Their data define novel in vivo mechanistic roles for ankyrin-G, implicate ankyrin-G as necessary for compensatory cardiac physiological remodeling under stress, and implicate disruption of ankyrin-G in the development and progression of human heart failure.

18.
Front Immunol ; 9: 561, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29632532

RESUMEN

Francisella tularensis is a remarkably infectious facultative intracellular bacterium of macrophages that causes tularemia. Early evasion of host immune responses contributes to the success of F. tularensis as a pathogen. F. tularensis entry into human monocytes and macrophages is mediated by the major phagocytic receptor, complement receptor 3 (CR3, CD11b/CD18). We recently determined that despite a significant increase in macrophage uptake following C3 opsonization of the virulent Type A F. tularensis spp. tularensis Schu S4, this phagocytic pathway results in limited pro-inflammatory cytokine production. Notably, MAP kinase/ERK activation is suppressed immediately during C3-opsonized Schu S4-CR3 phagocytosis. A mathematical model of CR3-TLR2 crosstalk predicted early involvement of Ras GTPase-activating protein (RasGAP) in immune suppression by CR3. Here, we link CR3-mediated uptake of opsonized Schu S4 by human monocytes and macrophages with inhibition of early signal 1 inflammasome activation, evidenced by limited caspase-1 cleavage and IL-18 release. This inhibition is due to increased RasGAP activity, leading to a reduction in the Ras-ERK signaling cascade upstream of the early inflammasome activation event. Thus, our data uncover a novel signaling pathway mediated by CR3 following engagement of opsonized virulent F. tularensis to limit inflammasome activation in human phagocytic cells, thereby contributing to evasion of the host innate immune system.


Asunto(s)
Francisella tularensis/inmunología , Inflamasomas/inmunología , Antígeno de Macrófago-1/inmunología , Macrófagos/inmunología , Fagocitosis/inmunología , Proteínas Activadoras de ras GTPasa/inmunología , Caspasa 1/inmunología , Caspasa 1/metabolismo , Células Cultivadas , Francisella tularensis/fisiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Evasión Inmune/inmunología , Inflamasomas/metabolismo , Interleucina-18/inmunología , Interleucina-18/metabolismo , Antígeno de Macrófago-1/metabolismo , Macrófagos/microbiología , Monocitos/inmunología , Monocitos/microbiología , Transducción de Señal/inmunología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo
19.
mBio ; 9(1)2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29295913

RESUMEN

The ability to grow at mammalian body temperatures is critical for pathogen infection of humans. For the thermally dimorphic fungal pathogen Histoplasma capsulatum, elevated temperature is required for differentiation of mycelia or conidia into yeast cells, a step critical for invasion and replication within phagocytic immune cells. Posttranslational glycosylation of extracellular proteins characterizes factors produced by the pathogenic yeast cells but not those of avirulent mycelia, correlating glycosylation with infection. Histoplasma yeast cells lacking the Pmt1 and Pmt2 protein mannosyltransferases, which catalyze O-linked mannosylation of proteins, are severely attenuated during infection of mammalian hosts. Cells lacking Pmt2 have altered surface characteristics that increase recognition of yeast cells by the macrophage mannose receptor and reduce recognition by the ß-glucan receptor Dectin-1. Despite these changes, yeast cells lacking these factors still associate with and survive within phagocytes. Depletion of macrophages or neutrophils in vivo does not recover the virulence of the mutant yeast cells. We show that yeast cells lacking Pmt functions are more sensitive to thermal stress in vitro and consequently are unable to productively infect mice, even in the absence of fever. Treatment of mice with cyclophosphamide reduces the normal core body temperature of mice, and this decrease is sufficient to restore the infectivity of O-mannosylation-deficient yeast cells. These findings demonstrate that O-mannosylation of proteins increases the thermotolerance of Histoplasma yeast cells, which facilitates infection of mammalian hosts.IMPORTANCE For dimorphic fungal pathogens, mammalian body temperature can have contrasting roles. Mammalian body temperature induces differentiation of the fungal pathogen Histoplasma capsulatum into a pathogenic state characterized by infection of host phagocytes. On the other hand, elevated temperatures represent a significant barrier to infection by many microbes. By functionally characterizing cells lacking O-linked mannosylation enzymes, we show that protein mannosylation confers thermotolerance on H. capsulatum, enabling infection of mammalian hosts.


Asunto(s)
Proteínas Fúngicas/metabolismo , Histoplasma/fisiología , Histoplasma/efectos de la radiación , Manosiltransferasas/metabolismo , Viabilidad Microbiana/efectos de la radiación , Procesamiento Proteico-Postraduccional , Animales , Modelos Animales de Enfermedad , Histoplasma/metabolismo , Histoplasmosis/microbiología , Histoplasmosis/patología , Ratones Endogámicos C57BL , Virulencia
20.
Cell Rep ; 21(1): 126-140, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28978467

RESUMEN

Despite its prominent role as a C-type lectin (CTL) pattern recognition receptor, mannose receptor (MR, CD206)-specific signaling molecules and pathways are unknown. The MR is highly expressed on human macrophages, regulating endocytosis, phagocytosis, and immune responses and mediating Mycobacterium tuberculosis (M.tb) phagocytosis by human macrophages, thereby limiting phagosome-lysosome (P-L) fusion. We identified human MR-associated proteins using phosphorylated and non-phosphorylated MR cytoplasmic tail peptides. We found that MR binds FcRγ-chain, which is required for MR plasma membrane localization and M.tb cell association. Additionally, we discovered that MR-mediated M.tb association triggers immediate MR tyrosine residue phosphorylation and Grb2 recruitment, activating the Rac/Pak/Cdc-42 signaling cascade important for M.tb uptake. MR activation subsequently recruits SHP-1 to the M.tb-containing phagosome, where its activity limits PI(3)P generation at the phagosome and M.tb P-L fusion and promotes M.tb growth. In sum, we identify human MR signaling pathways that temporally regulate phagocytosis and P-L fusion during M.tb infection.


Asunto(s)
Proteína Adaptadora GRB2/genética , Interacciones Huésped-Patógeno , Lectinas Tipo C/genética , Macrófagos/metabolismo , Lectinas de Unión a Manosa/genética , Mycobacterium tuberculosis/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Receptores de Superficie Celular/genética , Receptores de IgG/genética , Proteína Adaptadora GRB2/metabolismo , Regulación de la Expresión Génica , Humanos , Lectinas Tipo C/metabolismo , Lisosomas/metabolismo , Lisosomas/microbiología , Macrófagos/microbiología , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Fusión de Membrana , Mycobacterium tuberculosis/crecimiento & desarrollo , Fagocitosis/genética , Fagosomas/metabolismo , Fagosomas/microbiología , Fosforilación , Cultivo Primario de Células , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de IgG/metabolismo , Transducción de Señal , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/microbiología , Tirosina/metabolismo , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...