Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
EMBO J ; 33(9): 994-1010, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24431222

RESUMEN

There is emerging evidence that stem cells can rejuvenate damaged cells by mitochondrial transfer. Earlier studies show that epithelial mitochondrial dysfunction is critical in asthma pathogenesis. Here we show for the first time that Miro1, a mitochondrial Rho-GTPase, regulates intercellular mitochondrial movement from mesenchymal stem cells (MSC) to epithelial cells (EC). We demonstrate that overexpression of Miro1 in MSC (MSCmiro(Hi)) leads to enhanced mitochondrial transfer and rescue of epithelial injury, while Miro1 knockdown (MSCmiro(Lo)) leads to loss of efficacy. Treatment with MSCmiro(Hi) was associated with greater therapeutic efficacy, when compared to control MSC, in mouse models of rotenone (Rot) induced airway injury and allergic airway inflammation (AAI). Notably, airway hyperresponsiveness and remodeling were reversed by MSCmiro(Hi) in three separate allergen-induced asthma models. In a human in vitro system, MSCmiro(Hi) reversed mitochondrial dysfunction in bronchial epithelial cells treated with pro-inflammatory supernatant of IL-13-induced macrophages. Anti-inflammatory MSC products like NO, TGF-ß, IL-10 and PGE2, were unchanged by Miro1 overexpression, excluding non-specific paracrine effects. In summary, Miro1 overexpression leads to increased stem cell repair.


Asunto(s)
Lesión Pulmonar/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Mitocondrias/metabolismo , Proteínas de Unión al GTP rho/fisiología , Animales , Transporte Biológico/genética , Células Cultivadas , Terapia Genética/métodos , Humanos , Pulmón/patología , Lesión Pulmonar/patología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Mitocondrias/trasplante , Células 3T3 NIH , Nanotubos , Resultado del Tratamiento , Proteínas de Unión al GTP rho/genética
2.
J Biol Chem ; 287(14): 11108-21, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22337888

RESUMEN

With rising incidence of acquired drug resistance among life-threatening pathogens, alternative approaches to improve therapy and vaccination have taken center stage. To this end, genome-wide and pathway-specific siRNA libraries are being employed increasingly to identify genes that regulate immune responses against a number of pathogens. In this study using calcium and cysteine protease pathway-specific siRNA libraries, we identified genes that play critical roles in modulating diverse functions of dendritic cells (DCs) during Mycobacterium tuberculosis infection. Knockdown of many of these genes in the two pathways resulted in reduced bacterial burden within DCs. These included genes that regulated activation of transcription factors, ubiquitin-specific peptidases, and genes that are involved in autophagy and neddylation. Knockdown of certain genes increased the expression of IL-12p40 and surface densities of costimulatory molecules in an antigen- and receptor-specific manner. Increased IL-12p40 and costimulatory molecules on DCs also promoted the development of Th1 responses from a Th2 inducing antigen. Furthermore, modulation of autophagy and oxidative burst appeared to be one of the mechanisms by which these genes regulated survival of M. tuberculosis within DCs. Although some genes regulated specific responses, others regulated multiple responses that included IL-12 production, T cell priming, as well as intracellular survival of M. tuberculosis. Further dissection of the mechanisms such as neddylation, by which these genes regulate immune responses, would improve our understanding of host parameters that are modulated during M. tuberculosis infection.


Asunto(s)
Calcio/metabolismo , Proteasas de Cisteína/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Inmunidad/genética , Mycobacterium tuberculosis/fisiología , Animales , Autofagia/genética , Autofagia/inmunología , Calmodulina/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Subunidad p40 de la Interleucina-12/biosíntesis , Ratones , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Estallido Respiratorio/genética , Estallido Respiratorio/inmunología , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/microbiología , Receptor Toll-Like 2/metabolismo
3.
Biomol Concepts ; 11(1): 76-85, 2020 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-32271156

RESUMEN

Interferon-gamma (IFN-γ) is a key cytokine that mediates immunity to tuberculosis (TB). Mycobacterium tuberculosis (M. tb) is known to downregulate the surface expression of IFN-γ receptor (IFN-γR) on macrophages and peripheral blood mononuclear cells (PBMCs) of patients with active TB disease. Many M. tb antigens also downmodulate IFN-γR levels in macrophages when compared with healthy controls. In the current study, we aimed at deciphering key factors involved in M. tb mediated downregulation of IFN-γR levels on macrophage surface. Our data showed that both M. tb H37Rv and M. bovis BCG infections mediate downmodulation of IFN-γR on human macrophages. This downmodulation is regulated at the level of TLR signaling pathway, second messengers such as calcium and cellular kinases i.e. PKC and ERK-MAPK, indicating that fine tuning of calcium response is critical to maintaining IFN-γR levels on macrophage surface. In addition, genes in the calcium and cysteine protease pathways which were previously identified by us to play a negative role during M. tb infection, also regulated IFN-γR expression. Thus, modulations in IFN-γR levels by utilizing host machinery may be a key immune suppressive strategy adopted by the TB pathogen to ensure its persistence and thwart host defense.


Asunto(s)
Calcio/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Mycobacterium tuberculosis/fisiología , Receptores de Interferón/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Línea Celular , Citocinas/metabolismo , Endopeptidasas/genética , Endopeptidasas/metabolismo , Femenino , Homeostasis , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucocitos Mononucleares/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Endogámicos BALB C , Mycobacterium bovis/fisiología , Proteína Quinasa C/metabolismo , ARN Interferente Pequeño , Receptores de Interferón/genética , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Receptor de Interferón gamma
4.
Biomol Concepts ; 11(1): 230-239, 2020 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-33726488

RESUMEN

Staphylococcus aureus (S. aureus) is a gram-positive bacteria, which causes various fatal respiratory infections including pneumonia. The emergence of Methicillin-Resistance Staphylococcus aureus (MRSA) demands a thorough understanding of host-pathogen interactions. Here we report the role of calcium in regulating defence responses of S. aureus in macrophages. Regulating calcium fluxes in cells by different routes differentially governs the expression of T cell costimulatory molecule CD80 and Th1 promoting IL-12 receptor. Inhibiting calcium influx from extracellular medium increased expression of IFN-γ and IL-10 while blocking calcium release from the intracellular stores inhibited TGF-ß levels. Blocking voltage-gated calcium channels (VGCC) inhibited the expression of multiple cytokines. While VGCC regulated the expression of apoptosis protein Bax, extracellular calcium-regulated the expression of Cytochrome-C. Similarly, VGCC regulated the expression of autophagy initiator Beclin-1. Blocking VGCC or calcium release from intracellular stores promoted phagosome-lysosome fusion, while activating VGCC inhibited phagosomelysosome fusion. Finally, calcium homeostasis regulated intracellular growth of Staphylococcus, although using different mechanisms. While blocking extracellular calcium influx seems to rely on IFN-γ and IL-12Rß receptor mediated reduction in bacterial survival, blocking either intracellular calcium release or via VGCC route seem to rely on enhanced autophagy mediated reduction of intracellular bacterial survival. These results point to fine-tuning of defence responses by routes of calcium homeostasis.


Asunto(s)
Calcio/metabolismo , Macrófagos/metabolismo , Sustancias Protectoras/metabolismo , Staphylococcus aureus/crecimiento & desarrollo , Animales , Células Cultivadas , Citocinas/análisis , Citocinas/biosíntesis , Homeostasis , Ratones
5.
Biomol Concepts ; 9(1): 64-79, 2018 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-29856726

RESUMEN

A key player in driving cellular immunity, IFN-γ is capable of orchestrating numerous protective functions to heighten immune responses in infections and cancers. It can exhibit its immunomodulatory effects by enhancing antigen processing and presentation, increasing leukocyte trafficking, inducing an anti-viral state, boosting the anti-microbial functions and affecting cellular proliferation and apoptosis. A complex interplay between immune cell activity and IFN-γ through coordinated integration of signals from other pathways involving cytokines and Pattern Recognition Receptors (PRRs) such as Interleukin (IL)-4, TNF-α, Lipopolysaccharide (LPS), Type-I Interferons (IFNS) etc. leads to initiation of a cascade of pro-inflammatory responses. Microarray data has unraveled numerous genes whose transcriptional regulation is influenced by IFN-γ. Consequently, IFN-γ stimulated cells display altered expression of many such target genes which mediate its downstream effector functions. The importance of IFN-γ is further reinforced by the fact that mice possessing disruptions in the IFN-γ gene or its receptor develop extreme susceptibility to infectious diseases and rapidly succumb to them. In this review, we attempt to elucidate the biological functions and physiological importance of this versatile cytokine. The functional implications of its biological activity in several infectious diseases and autoimmune pathologies are also discussed. As a counter strategy, many virulent pathogenic species have devised ways to thwart IFN-γ endowed immune-protection. Thus, IFN-γ mediated host-pathogen interactions are critical for our understanding of disease mechanisms and these aspects also manifest enormous therapeutic importance for the annulment of various infections and autoimmune conditions.


Asunto(s)
Enfermedades Transmisibles/inmunología , Interferón gamma/fisiología , Animales , Enfermedades Transmisibles/tratamiento farmacológico , Enfermedades Transmisibles/metabolismo , Citocinas/fisiología , Regulación de la Expresión Génica , Humanos , Interferón gamma/metabolismo , Interferón gamma/uso terapéutico , Listeriosis/inmunología , Ratones , Infecciones por Mycobacterium/inmunología , Enfermedades Parasitarias/inmunología , Ratas , Infecciones por Salmonella/inmunología
6.
J Leukoc Biol ; 102(5): 1249-1259, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28877954

RESUMEN

Microorganisms are known to devise various strategies to thwart protective responses by the host. One such strategy is to incorporate sequences and domains in their genes/proteins that have similarity to various domains of the host proteins. In this study, we report that Mycobacterium tuberculosis protein Rv3529c exhibits significant similarity to the death domain of the TLR pathway adaptor protein MyD88. Incubation of macrophages with Rv3529c specifically inhibited TLR2-mediated proinflammatory responses. This included attenuated oxidative burst, reduced phosphorylation of MAPK-ERK, reduced activation of transcription factor NF-κB and reduced secretion of proinflammatory cytokines IFN-γ, IL-6, and IL-17A with a concomitant increased secretion of suppressor cytokines IL-10 and TGF-ß. Importantly, Rv3529c significantly inhibited TLR2-induced association of MyD88 with IRAK1 by competitively binding with IRAK1. Further, Rv3529c mediated inhibition of apoptosis and phagosome-lysosome fusion. Lastly, incubation of macrophages with Rv3529c increased bacterial burden inside macrophages. The data presented show another strategy evolved by M. tuberculosis toward immune evasion that centers on incorporating sequences in proteins that are similar to crucial proteins in the innate immune system of the host.


Asunto(s)
Proteínas Bacterianas/farmacología , Evasión Inmune , Macrófagos/microbiología , Mycobacterium tuberculosis/inmunología , Receptor Toll-Like 2/inmunología , Animales , Carga Bacteriana , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Regulación de la Expresión Génica , Interferón gamma/genética , Interferón gamma/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Lisosomas/efectos de los fármacos , Lisosomas/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Fusión de Membrana/efectos de los fármacos , Fusión de Membrana/inmunología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/inmunología , Imitación Molecular , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/patogenicidad , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Fagosomas/efectos de los fármacos , Fagosomas/inmunología , Cultivo Primario de Células , Dominios Proteicos , Estallido Respiratorio/inmunología , Transducción de Señal , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 2/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
7.
PLoS One ; 11(10): e0163845, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27723836

RESUMEN

The prevalence of Mycobacterium tuberculosis (M. tb) strains eliciting drug resistance has necessitated the need for understanding the complexities of host pathogen interactions. The regulation of calcium homeostasis by Voltage Gated Calcium Channel (VGCCs) upon M. tb infection has recently assumed importance in this area. We previously showed a suppressor role of VGCC during M. tb infections and recently reported the mechanisms of its regulation by M. tb. Here in this report, we further characterize the role of VGCC in mediating defence responses of macrophages during mycobacterial infection. We report that activation of VGCC during infection synergistically downmodulates the generation of oxidative burst (ROS) by macrophages. This attenuation of ROS is regulated in a manner which is dependent on Toll like Receptor (TLR) and also on the route of calcium influx, Protein Kinase C (PKC) and by Mitogen Activation Protein Kinase (MAPK) pathways. VGCC activation during infection increases cell survival and downmodulates autophagy. Concomitantly, pro-inflammatory responses such as IL-12 and IFN-γ secretion and the levels of their receptors on cell surface are inhibited. Finally, the ability of phagosomes to fuse with lysosomes in M. bovis BCG and M. tb H37Rv infected macrophages is also compromised when VGCC activation occurs during infection. The results point towards a well-orchestrated strategy adopted by mycobacteria to supress protective responses mounted by the host. This begins with the increase in the surface levels of VGCCs by mycobacteria and their antigens by well-controlled and regulated mechanisms. Subsequent activation of the upregulated VGCC following tweaking of calcium levels by molecular sensors in turn mediates suppressor responses and prepare the macrophages for long term persistent infection.


Asunto(s)
Canales de Calcio Tipo L/inmunología , Señalización del Calcio/inmunología , Macrófagos/inmunología , Infecciones por Mycobacterium/inmunología , Mycobacterium bovis/inmunología , Animales , Línea Celular Tumoral , Femenino , Humanos , Interferón gamma/inmunología , Interleucina-12/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Especies Reactivas de Oxígeno/inmunología
8.
PLoS One ; 10(4): e0124263, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25915405

RESUMEN

We demonstrated earlier the inhibitory role played by Voltage Gated Calcium Channels (VGCCs) in regulating Mycobacterium tuberculosis (M. tb) survival and pathogenesis. In this report, we investigated mechanisms and key players that regulate the surface expression of VGCC-CACNA1S by Rv2463 and M. tb infection in macrophages. Our earlier work identified Rv2463 to be expressed at early times post infection in macrophages that induced suppressor responses to dendritic cells and macrophages. Our results in this study demonstrate a role of MyD88 independent TLR pathway in mediating CACNA1S expression. Dissecting the role for second messengers, we show that calcium homeostasis plays a key role in CACNA1S expression during M. tb infection. Using siRNAs against molecular sensors of calcium regulation, we show an involvement of ER associated Stromal Interaction Molecules 1 and 2 (STIM1 and STIM2), and transcription factor pCREB, towards CACNA1S expression that also involved the MyD88 independent pathway. Interestingly, reactive oxygen species played a negative role in M. tb mediated CACNA1S expression. Further, a cross-regulation of ROS and pCREB was noted that governed CACNA1S expression. Characterizing the mechanisms governing CACNA1S expression would improve our understanding of the regulation of VGCC expression and its role in M. tb pathogenesis during M. tb infection.


Asunto(s)
Canales de Calcio/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Tuberculosis/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio Tipo L , Moléculas de Adhesión Celular/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Expresión Génica , Proteínas de la Membrana/metabolismo , Ratones , Modelos Biológicos , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas de Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Estallido Respiratorio/genética , Transducción de Señal , Molécula de Interacción Estromal 1 , Molécula de Interacción Estromal 2 , Factores de Transcripción/metabolismo , Tuberculosis/genética , Tuberculosis/microbiología
9.
PLoS One ; 10(7): e0131767, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26132135

RESUMEN

The emergence of drug resistant strains of Mycobacterium tuberculosis (M. tuberculosis) together with reports of co-infections with the human immunodeficiency virus (HIV) has renewed interest to better understand the intricate mechanisms prevalent during co-infections. In this study we report a synergistic effect of M. tuberculosis and HIV-1, and their antigens Rv3416 and Nef, respectively, in inhibiting apoptosis of macrophages. This inhibition involves the TLR2 pathway and second messengers that play complementing and contrasting roles in regulating apoptosis. Interestingly, the route of calcium influx into cells differentially regulates apoptosis during antigenic co-stimulation. While calcium released from intracellular stores was anti-apoptotic, calcium influx from the external milieu was pro-apoptotic. Further, molecular sensors of intracellular calcium release aid in antigen mediated inhibition of apoptosis. A cross-regulation between oxidative burst and differential routing of calcium influx governed apoptosis. Interestingly, the HIV-1 Nef supported anti-apoptotic responses in macrophages whereas Vpu had no significant effect. These results point to a synergistic liaison between M. tuberculosis and HIV-1 in regulating macrophage apoptosis.


Asunto(s)
Apoptosis , Calcio/metabolismo , VIH-1 , Macrófagos/metabolismo , Mycobacterium tuberculosis , Receptor Toll-Like 2/metabolismo , Antígenos/metabolismo , Proteínas Bacterianas/metabolismo , Células Cultivadas , Coinfección , Citocromos c/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Infecciones por VIH/complicaciones , Homeostasis , Humanos , Leucocitos Mononucleares/citología , Macrófagos/microbiología , Macrófagos/virología , Potencial de la Membrana Mitocondrial , ARN Interferente Pequeño/metabolismo , Estallido Respiratorio , Transducción de Señal , Tuberculosis/complicaciones , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
10.
PLoS One ; 9(5): e96427, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24797940

RESUMEN

Our previous work has demonstrated the roles played by L-type Voltage Gated Calcium Channels (VGCC) in regulating Mycobacterium tuberculosis (M. tb) survival and pathogenesis. Here we decipher mechanisms and pathways engaged by the pathogen to regulate VGCC expression in macrophages. We show that M. tb and its antigen Rv3416 use phospho-CREB (pCREB), Reactive Oxygen Species (ROS), Protein Kinase C (PKC) and Mitogen Activated Protein Kinase (MAPK) to modulate VGCC expression in macrophages. siRNA mediated knockdown of MyD88, IRAK1, IRAK2 or TRAF6 significantly inhibited antigen mediated VGCC expression. Inhibiting Protein Kinase C (PKC) or MEK-ERK1/2 further increased VGCC expression. Interestingly, inhibiting intracellular calcium release upregulated antigen mediated VGCC expression, while inhibiting extracellular calcium influx had no significant effect. siRNA mediated knockdown of transcription factors c-Jun, SOX5 and CREB significantly inhibited Rv3416 mediated VGCC expression. A dynamic reciprocal cross-regulation between ROS and pCREB was observed that in turn governed VGCC expression with ROS playing a limiting role in the process. Further dissection of the mechanisms such as the interplay between ROS and pCREB would improve our understanding of the regulation of VGCC expression during M. tb infection.


Asunto(s)
Canales de Calcio/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Mycobacterium tuberculosis/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Antígenos Bacterianos/fisiología , Línea Celular , Humanos , Macrófagos/metabolismo , Redes y Vías Metabólicas/inmunología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/fisiología , Mycobacterium tuberculosis/patogenicidad , Proteína Quinasa C/metabolismo , Proteína Quinasa C/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA