Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nat Immunol ; 11(8): 751-8, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20622882

RESUMO

The fate of infected macrophages has an essential role in protection against Mycobacterium tuberculosis by regulating innate and adaptive immunity. M. tuberculosis exploits cell necrosis to exit from macrophages and spread. In contrast, apoptosis, which is characterized by an intact plasma membrane, is an innate mechanism that results in lower bacterial viability. Virulent M. tuberculosis inhibits apoptosis and promotes necrotic cell death by inhibiting production of prostaglandin E(2). Here we show that by activating the 5-lipoxygenase pathway, M. tuberculosis not only inhibited apoptosis but also prevented cross-presentation of its antigens by dendritic cells, which impeded the initiation of T cell immunity. Our results explain why T cell priming in response to M. tuberculosis is delayed and emphasize the importance of early immunity.


Assuntos
Dinoprostona/antagonistas & inibidores , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Imunidade Adaptativa/imunologia , Animais , Apresentação de Antígeno , Antígenos de Bactérias/imunologia , Apoptose/imunologia , Araquidonato 5-Lipoxigenase/imunologia , Araquidonato 5-Lipoxigenase/metabolismo , Células Dendríticas/imunologia , Dinoprostona/biossíntese , Dinoprostona/imunologia , Modelos Animais de Doenças , Ativação Enzimática , Imunidade Inata/imunologia , Inibidores de Lipoxigenase , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Necrose , Transdução de Sinais , Tuberculose/metabolismo , Tuberculose/microbiologia , Tuberculose/patologia , Virulência
2.
Nat Immunol ; 10(8): 899-906, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19561612

RESUMO

Induction of macrophage necrosis is a strategy used by virulent Mycobacterium tuberculosis (Mtb) to avoid innate host defense. In contrast, attenuated Mtb causes apoptosis, which limits bacterial replication and promotes T cell cross-priming by antigen-presenting cells. Here we show that Mtb infection causes plasma membrane microdisruptions. Resealing of these lesions, a process crucial for preventing necrosis and promoting apoptosis, required translocation of lysosomal and Golgi apparatus-derived vesicles to the plasma membrane. Plasma membrane repair depended on prostaglandin E(2) (PGE(2)), which regulates synaptotagmin 7 (Syt-7), the calcium sensor involved in the lysosome-mediated repair mechanism. By inducing production of lipoxin A(4) (LXA(4)), which blocks PGE(2) biosynthesis, virulent Mtb prevented membrane repair and induced necrosis. Thus, virulent Mtb impairs macrophage plasma membrane repair to evade host defenses.


Assuntos
Membrana Celular/patologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/fisiologia , Animais , Apoptose , Membrana Celular/imunologia , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Células Cultivadas , Dinoprostona/metabolismo , Complexo de Golgi/fisiologia , Humanos , Lipoxinas/metabolismo , Lisossomos/fisiologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Necrose , Sinaptotagminas/metabolismo , Virulência
3.
Nat Immunol ; 9(10): 1189-97, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18794848

RESUMO

Macrophages infected with attenuated Mycobacterium tuberculosis strain H37Ra become apoptotic, which limits bacterial replication and facilitates antigen presentation. Here we demonstrate that cells infected with H37Ra became apoptotic after the formation of an apoptotic envelope on their surface was complete. This process required exposure of phosphatidylserine on the cell surface, followed by deposition of the phospholipid-binding protein annexin-1 and then transglutaminase-mediated crosslinking of annexin-1 through its amino-terminal domain. In macrophages infected with the virulent strain H37Rv, in contrast, the amino-terminal domain of annexin-1 was removed by proteolysis, thus preventing completion of the apoptotic envelope, which resulted in macrophage death by necrosis. Virulent M. tuberculosis therefore avoids the host defense system by blocking formation of the apoptotic envelope, which leads to macrophage necrosis and dissemination of infection in the lung.


Assuntos
Anexinas/metabolismo , Apoptose/imunologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Tuberculose/imunologia , Animais , Anexinas/imunologia , Humanos , Immunoblotting , Macrófagos/imunologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Necrose/imunologia , Inibidor 2 de Ativador de Plasminogênio/imunologia , Inibidor 2 de Ativador de Plasminogênio/metabolismo , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Virulência
4.
J Immunol ; 200(9): 3160-3169, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29592961

RESUMO

Mycobacterium tuberculosis is an extremely successful pathogen, and its success is widely attributed to its ability to manipulate the intracellular environment of macrophages. A central phenomenon of tuberculosis pathology enabling immune evasion is the capacity of virulent M. tuberculosis (H37Rv) to induce macrophage necrosis, which facilitates the escape of the mycobacteria from the macrophage and spread of infection. In contrast, avirulent M. tuberculosis (H37Ra) induces macrophage apoptosis, which permits Ag presentation and activation of adaptive immunity. Previously, we found that H37Rv induces plasma membrane microdisruptions, leading to necrosis in the absence of plasma membrane repair. In contrast, H37Ra permits plasma membrane repair, which changes the host cell death modality to apoptosis, suggesting that membrane repair is critical for sequestering the pathogen in apoptotic vesicles. However, mechanisms of plasma membrane repair induced in response to M. tuberculosis infection remain unknown. Plasma membrane repair is known to induce a Ca2+-mediated signaling, which recruits lysosomes to the area of damaged plasma membrane sites for its resealing. In this study, we found that the small GTPase Arl8b is required for plasma membrane repair by controlling the exocytosis of lysosomes in cell lines and in human primary macrophages. Importantly, we found that the Arl8b secretion pathway is crucial to control the type of cell death of the M. tuberculosis-infected macrophages. Indeed, Arl8b-depleted macrophages infected with avirulent H37Ra undergo necrotic instead of apoptotic cell death. These findings suggest that membrane repair mediated by Arl8b may be an important mechanism distinguishing avirulent from virulent M. tuberculosis-induced necrotic cell death.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Membrana Celular/metabolismo , Lisossomos/metabolismo , Macrófagos/microbiologia , Tuberculose/metabolismo , Apoptose/fisiologia , Humanos , Evasão da Resposta Imune/fisiologia , Macrófagos/metabolismo , Mycobacterium tuberculosis/patogenicidade , Necrose/metabolismo , Necrose/microbiologia , Virulência/fisiologia
5.
J Immunol ; 190(8): 4196-204, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23487424

RESUMO

In vivo control of Mycobacterium tuberculosis reflects the balance between host immunity and bacterial evasion strategies. Effector Th1 cells that mediate protective immunity by depriving the bacterium of its intracellular niche are regulated to prevent overexuberant inflammation. One key immunoregulatory molecule is Tim3. Although Tim3 is generally recognized to downregulate Th1 responses, we recently described that its interaction with Galectin-9 expressed by M. tuberculosis-infected macrophages stimulates IL-1ß secretion, which is essential for survival in the mouse model. Why IL-1ß is required for host resistance to M. tuberculosis infection is unknown. In this article, we show that IL-1ß directly kills M. tuberculosis in murine and human macrophages and does so through the recruitment of other antimicrobial effector molecules. IL-1ß directly augments TNF signaling in macrophages through the upregulation of TNF secretion and TNFR1 cell surface expression, and results in activation of caspase-3. Thus, IL-1ß and downstream TNF production lead to caspase-dependent restriction of intracellular M. tuberculosis growth.


Assuntos
Caspase 3/metabolismo , Interleucina-1beta/fisiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/imunologia , Regulação para Cima , Animais , Células Cultivadas , Ativação Enzimática/imunologia , Humanos , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium tuberculosis/imunologia , Receptores do Fator de Necrose Tumoral/fisiologia , Regulação para Cima/imunologia
6.
FASEB J ; 27(9): 3827-36, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23759445

RESUMO

Prostaglandin E2 (PGE2) is an important biological mediator involved in the defense against Mycobacterium tuberculosis (Mtb) infection. Previously, we reported that in macrophages (Mϕs), infection with avirulent Mtb H37Ra resulted in inhibition of necrosis by an inhibitory effect on mitochondrial permeability transition via the PGE2 receptor EP2. However, human Mϕs also express EP4, a PGE2 receptor functionally closely related to EP2 that also couples to stimulatory guanine nucleotide binding protein, but the functional differences between EP2 and EP4 in Mtb-infected Mϕs have been unclear. EP4 antagonist addition to H37Ra-infected Mϕs inhibited the expression of cyclooxygenase 2 (COX2) and microsomal prostaglandin E synthase-1 (mPGES-1), which are involved in PGE2 production. Moreover, H37Ra infection induced PGE2 production through the Toll-like receptor (TLR) 2/p38 mitogen-activated protein kinase (MAPK) signaling pathway. Induction of COX2 and mPGES-1 expression by TLR2 stimulation or Mtb infection was increased after additional stimulation with EP4 agonist. Hence, in Mtb-infected Mϕs, PGE2 production induced by pathogen recognition receptors/p38 MAPK signaling is up-regulated by EP4-triggered signaling to maintain an effective PGE2 concentration.


Assuntos
Dinoprostona/metabolismo , Macrófagos/microbiologia , Mycobacterium tuberculosis/patogenicidade , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Humanos , Immunoblotting , Receptores de Prostaglandina E Subtipo EP4/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética
7.
mBio ; 12(1)2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33531400

RESUMO

Mycobacterium tuberculosis induces metabolic reprogramming in macrophages like the Warburg effect. This enhances antimicrobial performance at the expense of increased inflammation, which may promote a pathogen-permissive host environment. Since the NAD+-dependent protein deacetylase Sirtuin 3 (SIRT3) is an important regulator of mitochondrial metabolism and cellular redox homeostasis, we hypothesized that SIRT3 modulation mediates M. tuberculosis-induced metabolic reprogramming. Infection of immortalized and primary murine macrophages resulted in reduced levels of SIRT3 mRNA and protein and perturbation of SIRT3-regulated enzymes in the tricarboxylic acid cycle, electron transport chain, and glycolytic pathway. These changes were associated with increased reactive oxygen species and reduced antioxidant scavenging, thereby triggering mitochondrial stress and macrophage cell death. Relevance to tuberculosis disease in vivo was indicated by greater bacterial burden and immune pathology in M. tuberculosis-infected Sirt3-/- mice. CD11b+ lung leukocytes isolated from infected Sirt3-/- mice showed decreased levels of enzymes involved in central mitochondrial metabolic pathways, along with increased reactive oxygen species. Bacterial burden was also greater in lungs of LysMcreSirt3L2/L2 mice, demonstrating the importance of macrophage-specific SIRT3 after infection. These results support the model of SIRT3 as a major upstream regulatory factor, leading to metabolic reprogramming in macrophages by M. tuberculosisIMPORTANCE Tuberculosis, the disease caused by the bacterium M. tuberculosis, remains one of the top 10 causes of death worldwide. Macrophages, the first cells to encounter M. tuberculosis and critical for defense against infection, are hijacked by M. tuberculosis as a protected growth niche. M. tuberculosis-infected macrophages undergo metabolic reprogramming where key mitochondrial pathways are modulated, but the mechanisms driving this metabolic shift is unknown. Our study demonstrates that M. tuberculosis downregulates Sirtuin 3 (SIRT3), an important regulator of mitochondrial metabolism, leading to SIRT3-dependent transcriptional downregulation of mitochondrial metabolic proteins, which is followed by oxidative stress and macrophage necrosis. This study identifies SIRT3 modulation as a key event in M. tuberculosis-induced metabolic reprograming in macrophages that defend against tuberculosis.


Assuntos
Macrófagos/metabolismo , Mitocôndrias/metabolismo , Mycobacterium tuberculosis/patogenicidade , Sirtuína 3/fisiologia , Animais , Reprogramação Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Receptor 2 Toll-Like/fisiologia , Receptor 4 Toll-Like/fisiologia
8.
Cell Host Microbe ; 12(3): 289-300, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-22980326

RESUMO

Mycobacterium tuberculosis persists within macrophages in an arrested phagosome and depends upon necrosis to elude immunity and disseminate. Although apoptosis of M. tuberculosis-infected macrophages is associated with reduced bacterial growth, the bacteria are relatively resistant to other forms of death, leaving the mechanism underlying this observation unresolved. We find that after apoptosis, M. tuberculosis-infected macrophages are rapidly taken up by uninfected macrophages through efferocytosis, a dedicated apoptotic cell engulfment process. Efferocytosis of M. tuberculosis sequestered within an apoptotic macrophage further compartmentalizes the bacterium and delivers it along with the apoptotic cell debris to the lysosomal compartment. M. tuberculosis is killed only after efferocytosis, indicating that apoptosis itself is not intrinsically bactericidal but requires subsequent phagocytic uptake and lysosomal fusion of the apoptotic body harboring the bacterium. While efferocytosis is recognized as a constitutive housekeeping function of macrophages, these data indicate that it can also function as an antimicrobial effector mechanism.


Assuntos
Apoptose , Macrófagos/imunologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Fagocitose , Animais , Células Cultivadas , Evasão da Resposta Imune , Lisossomos/metabolismo , Lisossomos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência
9.
Microbes Infect ; 13(8-9): 749-56, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21458584

RESUMO

Eicosanoids regulate whether human and murine macrophages infected with Mycobacterium tuberculosis die by apoptosis or necrosis. The death modality is important since apoptosis is associated with diminished pathogen viability and should be viewed as a form of innate immunity. Apoptotic vesicles derived from infected macrophages are also an important source of bacterial antigens that can be acquired by dendritic cells to prime antigen-specific T cells. This review integrates in vitro and in vivo data on how apoptosis of infected macrophages is linked to development of T cell immunity against M. tuberculosis.


Assuntos
Apoptose/imunologia , Eicosanoides/imunologia , Tuberculose/imunologia , Animais , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Celular/imunologia , Macrófagos/imunologia , Camundongos , Mycobacterium tuberculosis/imunologia
10.
Nat Rev Microbiol ; 8(9): 668-74, 2010 09.
Artigo em Inglês | MEDLINE | ID: mdl-20676146

RESUMO

Virulent Mycobacterium tuberculosis inhibits apoptosis and triggers necrosis of host macrophages to evade innate immunity and delay the initiation of adaptive immunity. By contrast, attenuated M. tuberculosis induces macrophage apoptosis, an innate defence mechanism that reduces bacterial viability. In this Opinion article, we describe how virulent M. tuberculosis blocks production of the eicosanoid lipid mediator prostaglandin E(2) (PGE(2)). PGE(2) production by infected macrophages prevents mitochondrial damage and initiates plasma membrane repair, two processes that are crucial for preventing necrosis and inducing apoptosis. Thus, M. tuberculosis-mediated modulation of eicosanoid production determines the death modality of the infected macrophage, which in turn has a substantial impact on the outcome of infection.


Assuntos
Apoptose , Evasão da Resposta Imune , Mycobacterium tuberculosis/imunologia , Animais , Eicosanoides/metabolismo , Humanos , Imunidade Inata , Macrófagos/citologia , Macrófagos/microbiologia
11.
J Exp Med ; 205(12): 2791-801, 2008 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-18955568

RESUMO

Virulent Mycobacterium tuberculosis (Mtb) induces a maladaptive cytolytic death modality, necrosis, which is advantageous for the pathogen. We report that necrosis of macrophages infected with the virulent Mtb strains H37Rv and Erdmann depends on predominant LXA(4) production that is part of the antiinflammatory and inflammation-resolving action induced by Mtb. Infection of macrophages with the avirulent H37Ra triggers production of high levels of the prostanoid PGE(2), which promotes protection against mitochondrial inner membrane perturbation and necrosis. In contrast to H37Ra infection, PGE(2) production is significantly reduced in H37Rv-infected macrophages. PGE(2) acts by engaging the PGE(2) receptor EP2, which induces cyclic AMP production and protein kinase A activation. To verify a role for PGE(2) in control of bacterial growth, we show that infection of prostaglandin E synthase (PGES)(-/-) macrophages in vitro with H37Rv resulted in significantly higher bacterial burden compared with wild-type macrophages. More importantly, PGES(-/-) mice harbor significantly higher Mtb lung burden 5 wk after low-dose aerosol infection with virulent Mtb. These in vitro and in vivo data indicate that PGE(2) plays a critical role in inhibition of Mtb replication.


Assuntos
Morte Celular/imunologia , Dinoprostona/imunologia , Imunidade Inata/fisiologia , Lipoxinas/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis , Receptores de Prostaglandina E/imunologia , Animais , Células Cultivadas , Dinoprostona/química , Dinoprostona/genética , Humanos , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/imunologia , Lipoxinas/química , Lipoxinas/genética , Pulmão/imunologia , Pulmão/microbiologia , Macrófagos/citologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Estrutura Molecular , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Prostaglandina-E Sintases , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E Subtipo EP2 , Tuberculose/imunologia
12.
J Immunol ; 176(6): 3707-16, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16517739

RESUMO

Infection of human monocyte-derived macrophages with Mycobacterium tuberculosis at low multiplicities of infection leads 48-72 h after the infection to cell death with the characteristics of apoptosis or necrosis. Predominant induction of one or the other cell death modality depends on differences in mitochondrial membrane perturbation induced by attenuated and virulent strains. Infection of macrophages with the attenuated H37Ra or the virulent H37Rv causes mitochondrial outer membrane permeabilization characterized by cytochrome c release from the mitochondrial intermembrane space and apoptosis. Mitochondrial outer membrane permeabilization is transient, peaks 6 h after infection, and requires Ca(2+) flux and B cell chronic lymphocytic leukemia/lymphoma 2-associated protein X translocation into mitochondria. In contrast, only the virulent H37Rv induces significant mitochondrial transmembrane potential (Deltapsi(m)) loss caused by mitochondrial permeability transition. Dissipation of Deltapsi(m) also peaks at 6 h after infection, is transient, is inhibited by the classical mitochondrial permeability transition inhibitor cyclosporine A, has a requirement for mitochondrial Ca(2+) loading, and is independent of B cell chronic lymphocytic leukemia/lymphoma translocation into the mitochondria. Transient dissipation of Deltapsi(m) 6 h after infection is essential for the induction of macrophage necrosis by Mtb, a mechanism that allows further dissemination of the pathogen and development of the disease.


Assuntos
Macrófagos/microbiologia , Macrófagos/patologia , Mitocôndrias/patologia , Membranas Mitocondriais/patologia , Mycobacterium tuberculosis/classificação , Mycobacterium tuberculosis/patogenicidade , Cálcio/química , Cálcio/metabolismo , Cátions Bivalentes/química , Permeabilidade da Membrana Celular , Células Cultivadas , Citocromos c/metabolismo , Humanos , Leucemia Linfocítica Crônica de Células B , Macrófagos/metabolismo , Microscopia Eletrônica , Mitocôndrias/metabolismo , Mitocôndrias/microbiologia , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/microbiologia , Necrose , Virulência , Proteína X Associada a bcl-2/metabolismo
13.
J Immunol ; 176(7): 4267-74, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16547264

RESUMO

We previously reported that macrophage exposure to attenuated strains of pathogenic mycobacteria at multiplicities of infection (MOI) < or = 10 triggers TNF-alpha-mediated apoptosis which reduces the viability of intracellular bacilli. Virulent strains were found to suppress macrophage apoptosis, and it was proposed that apoptosis is an innate defense against intracellular Mycobacterium tuberculosis analogous to apoptosis of virus-infected cells. The potential similarity of host cell responses to intracellular infection with mycobacteria and viruses suggests that M. tuberculosis might lyse infected macrophage when that niche is no longer needed. To investigate this question, we challenged murine macrophages with high intracellular bacillary loads. A sharp increase in cytolysis within 24 h was observed at MOI > or = 25. The primary death mode was apoptosis, based on nuclear morphology and phosphatidyl serine exposure, although the apoptotic cells progressed rapidly to necrosis. Apoptosis at high MOI differs markedly from low MOI apoptosis: it is potently induced by virulent M. tuberculosis, it is TNF-alpha-independent, and it does not reduce mycobacterial viability. Caspase inhibitors failed to prevent high MOI apoptosis, and macrophages deficient in caspase-3, MyD88, or TLR4 were equally susceptible as wild type. Apoptosis was reduced in the presence of cathepsin inhibitors, suggesting the involvement of lysosomal proteases in this novel death response. We conclude that the presence of high numbers of intracellular M. tuberculosis bacilli triggers a macrophage cell death pathway that could promote extracellular spread of infection and contribute to the formation of necrotic lesions in tuberculosis.


Assuntos
Apoptose , Macrófagos/citologia , Mycobacterium tuberculosis/fisiologia , Transdução de Sinais , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Caspase 3 , Inibidores de Caspase , Caspases/metabolismo , Catepsinas/antagonistas & inibidores , Catepsinas/metabolismo , Células Cultivadas , Quelantes/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/imunologia , Camundongos , Mycobacterium tuberculosis/imunologia , Fagócitos/citologia , Receptores Toll-Like/metabolismo
14.
J Infect Dis ; 191(8): 1292-300, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15776376

RESUMO

Infection of human macrophages with Mycobacterium tuberculosis leads to cell death that, depending on the M. tuberculosis strain, time course, and multiplicity of infection, may have predominant features of apoptosis or necrosis. A key feature of infection-induced necrosis is mitochondrial damage characterized by an irreversible increase in the mitochondrial permeability transition (MPT), which is associated with increased release of cytochrome c from the mitochondria and uncontrolled mycobacterial replication. In contrast, protection of the mitochondria from MPT favors apoptosis of M. tuberculosis-infected macrophages. Apoptosis of M. tuberculosis-infected macrophages is associated with killing of intracellular M. tuberculosis, and this may be enhanced when MPT is stabilized. Here, we show that cyclosporin A (CsA), an inhibitor of MPT, protects the mitochondria from release of cytochrome c and promotes the antimycobacterial activity of macrophages infected with M. tuberculosis H37Ra. Signaling by purinergic P2 receptors has previously been linked to the antimycobacterial activity of macrophages. In the present study, we found that infection with H37Ra inhibits P2X7 receptor (P2XR) signals and that CsA restores P2XR function in infected macrophages. Together, these data demonstrate that CsA promotes at least 2 antimycobacterial pathways of macrophages.


Assuntos
Macrófagos Alveolares/citologia , Macrófagos Alveolares/imunologia , Potenciais da Membrana/fisiologia , Mitocôndrias/fisiologia , Mycobacterium tuberculosis/imunologia , Trifosfato de Adenosina/metabolismo , Sobrevivência Celular , Ciclosporina/farmacologia , Citocromos c/metabolismo , Humanos , Macrófagos Alveolares/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/fisiologia , Necrose , Receptores Purinérgicos P2/metabolismo
15.
J Immunol ; 169(9): 5181-7, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12391235

RESUMO

Human macrophages (Mphi) respond to Mycobacterium tuberculosis (Mtb) infection by undergoing apoptosis, a cornerstone of effective antimycobacterial host defense. Virulent mycobacteria override this reaction by inducing necrosis leading to uncontrolled Mtb replication. Accordingly, Mphi death induced by inoculation with Mtb had the characteristics of apoptosis and necrosis and correlated with moderate increase of mitochondrial permeability transition (MPT), mitochondrial cytochrome c release, and caspase-9 and -3 activation. We hypothesized that changes in intramitochondrial Ca(2+) concentration ([Ca(2+)](m)) determine whether Mphi undergo either apoptosis or necrosis. Therefore, we induced mechanism(s) leading to predominant apoptosis or necrosis by modulating [Ca(2+)](m) and examined their physiological consequences. Adding calcium ionophore A23187 to Mphi inoculated with Mtb further increased calcium flux into the cells which is thought to lead to increased [Ca(2+)](m), blocked necrosis, stabilized MPT, decreased mitochondrial cytochrome c release, lowered caspase activation, and accompanied effective antimycobacterial activity. In contrast, Mphi infected with Mtb in presence of the mitochondrial calcium uniporter inhibitor ruthenium red showed increased mitochondrial swelling and cytochrome c release and decreased MPT and antimycobacterial activity. Thus, in Mtb-infected Mphi, high levels of mitochondrial membrane integrity, low levels of caspase activation, and diminished mitochondrial cytochrome c release are hallmarks of apoptosis and effective antimycobacterial activity. In contrast, breakdown of mitochondrial membrane integrity and increased caspase activation are characteristic of necrosis and uncontrolled Mtb replication.


Assuntos
Macrófagos/microbiologia , Mitocôndrias/microbiologia , Dilatação Mitocondrial/imunologia , Mycobacterium tuberculosis/imunologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Atividade Bactericida do Sangue/efeitos dos fármacos , Calcimicina/farmacologia , Canais de Cálcio , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Inibidores de Caspase , Caspases/metabolismo , Células Cultivadas , Grupo dos Citocromos c/antagonistas & inibidores , Grupo dos Citocromos c/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/microbiologia , Membranas Intracelulares/ultraestrutura , Ionóforos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/ultraestrutura , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/imunologia , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Dilatação Mitocondrial/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Necrose , Permeabilidade/efeitos dos fármacos , Rutênio Vermelho/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA