Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Front Immunol ; 14: 1244071, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37662946

RESUMEN

During Trypanosoma cruzi infection, macrophages phagocytose parasites and remove apoptotic cells through efferocytosis. While macrophage 1 (M1) produces proinflammatory cytokines and NO and fights infection, M2 macrophages are permissive host cells that express arginase 1 and play a role in tissue repair. The regulation of M1 and M2 phenotypes might either induce or impair macrophage-mediated immunity towards parasite control or persistence in chronic Chagas disease. Here, we highlight a key role of macrophage activation in early immune responses to T. cruzi that prevent escalating parasitemia, heart parasitism, and mortality during acute infection. We will discuss the mechanisms of macrophage activation and deactivation, such as T cell cytokines and efferocytosis, and how to improve macrophage-mediated immunity to prevent parasite persistence, inflammation, and the development of chagasic cardiomyopathy. Potential vaccines or therapy must enhance early T cell-macrophage crosstalk and parasite control to restrain the pathogenic outcomes of parasite-induced inflammation in the heart.


Asunto(s)
Enfermedad de Chagas , Macrófagos , Humanos , Citocinas , Inflamación , Apoptosis
2.
Commun Biol ; 5(1): 1421, 2022 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-36581764

RESUMEN

Adaptive immunity controls Trypanosoma cruzi infection, but the protozoan parasite persists and causes Chagas disease. T cells undergo apoptosis, and the efferocytosis of apoptotic cells might suppress macrophages and exacerbate parasite infection. Nonetheless, the receptors involved in the efferocytosis of apoptotic lymphocytes during infection remain unknow. Macrophages phagocytose apoptotic cells by using the TAM (Tyro3, Axl, Mer) family of receptors. To address how the efferocytosis of apoptotic cells affects macrophage-mediated immunity, we employ here Axl receptor- and Mer receptor-deficient mouse strains. In bone marrow-derived macrophages (BMDMs), both Axl and Mer receptors play a role in the efferocytosis of proapoptotic T cells from T. cruzi-infected mice. Moreover, treatment with a TAM receptor inhibitor blocks efferocytosis and upregulates M1 hallmarks induced by immune T cells from infected mice. Remarkably, the use of Axl-/- but not Mer-/- macrophages increases T-cell-induced M1 responses, such as nitric oxide production and control of parasite infection. Furthermore, infected Axl-/- mice show reduced peak parasitemia, defective efferocytosis, improved M1 responses, and ameliorated cardiac inflammation and fibrosis. Therefore, Axl induces efferocytosis, disrupts M1 responses, and promotes parasite infection and pathology in experimental Chagas disease. Axl stands as a potential host-direct target for switching macrophage phenotypes in infectious diseases.


Asunto(s)
Tirosina Quinasa del Receptor Axl , Enfermedad de Chagas , Macrófagos , Miocardio , Animales , Ratones , Proteínas Portadoras , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/patología , Fagocitosis , Ratones Noqueados , Tirosina Quinasa del Receptor Axl/genética , Corazón/parasitología , Miocardio/patología
3.
Front Immunol ; 12: 704429, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34249011

RESUMEN

In the innate immunity to Leishmania infection tissue-resident macrophages and inflammatory monocytes accumulate host-cell, effector, and efferocytosis functions. In addition, neutrophils, as host, effector, and apoptotic cells, as well as tissue-resident and monocyte-derived dendritic cells (DCs) imprint innate and adaptive immunity to Leishmania parasites. Macrophages develop phenotypes ranging from antimicrobial M1 to parasite-permissive M2, depending on mouse strain, Leishmania species, and T-cell cytokines. The Th1 (IFN-γ) and Th2 (IL-4) cytokines, which induce classically-activated (M1) or alternatively-activated (M2) macrophages, underlie resistance versus susceptibility to leishmaniasis. While macrophage phenotypes have been well discussed, new developments addressed the monocyte functional phenotypes in Leishmania infection. Here, we will emphasize the role of inflammatory monocytes to access how potential host-directed therapies for leishmaniasis, such as all-trans-retinoic acid (ATRA) and the ligand of Receptor Activator of Nuclear Factor-Kappa B (RANKL) might modulate immunity to Leishmania infection, by directly targeting monocytes to develop M1 or M2 phenotypes.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Leishmania/inmunología , Leishmaniasis/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Animales , Humanos , Macrófagos/parasitología , Ratones , Monocitos/parasitología
4.
Front Immunol ; 11: 886, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32477357

RESUMEN

Macrophages host Leishmania major infection, which causes cutaneous Leishmaniasis in humans. In the murine model, resistance to infection depends on the host immunity mediated by CD4 T-cell cytokines and macrophages. In association to other stimuli, the Th1 cytokine IFN-γ induces NO-mediated microbial killing by M1/classically-activated macrophages. By contrast, the Th2 cytokine IL-4 promotes M2/alternatively activated macrophages, which express arginase-1 and shelter infection. Other cytokines, such as RANKL, might also participate in the crosstalk between T cells and macrophages to restrict parasite infection. RANKL and its receptor RANK are known to play an essential role in bone remodeling, by inducing osteoclatogenesis. It has also been shown that RANKL stimulates antigen-presenting cells, such as DCs and macrophages, to enhance T cell responses. Here we investigated how RANKL directly modulates the effector macrophage phenotypes and immunity to L. major parasites. We found that inflammatory peritoneal macrophages from B6 mice express RANK and M2 features, such as CD301 (MGL) and CD206 (mannose receptor). Nonetheless, treatment with RANKL or IFN-γ induced macrophage differentiation into more mature F40/80hi macrophages able to produce IL-12 and TNF-α. In parallel, macrophages treated with RANKL, IFN-γ, or RANKL along with IFN-γ progressively downregulated the expression of the M2 hallmarks MGL, arginase-1, and CCL17. Moreover, a synergism between IFN-γ and RANKL enhanced inducible NO synthase (iNOS) expression and NO production by macrophages. These results are consistent with the idea that RANKL helps IFN-γ to induce a M2-like to M1 phenotype shift. Accordingly, concomitant treatment with RANKL and IFN-γ promoted macrophage-mediated immunity to L. major, by inducing NO and ROS-dependent parasite killing. Furthermore, by cooperating with IFN-γ, endogenous RANKL engages CD4 T-cell help toward L. major-infected macrophages to upregulate M1 and Th1 cytokine responses. Therefore, RANKL, in combination with IFN-γ, is a potential local therapeutic tool to improve immune responses in Leishmaniasis, by skewing M2-like into effector M1 macrophages.


Asunto(s)
Diferenciación Celular/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/parasitología , Ligando RANK/inmunología , Animales , Leishmania major , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal
5.
Front Immunol ; 9: 1569, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30038622

RESUMEN

Trypanosoma cruzi infects and replicates within a wide variety of immune and non-immune cells. Here, we investigated early cellular responses induced in NIH-3T3 fibroblasts upon infection with trypomastigote forms of T. cruzi. We show that fibroblasts were susceptible to T. cruzi infection and started to release trypomastigotes to the culture medium after 4 days of infection. Also, we found that T. cruzi infection reduced the number of fibroblasts in 3-day cell cultures, by altering fibroblast proliferation. Infected fibroblasts displayed distinctive phenotypic alterations, including enlarged and flattened morphology with a nuclei accumulation of senescence-associated heterochromatin foci. In addition, infection induced an overexpression of the enzyme senescence-associated ß-galactosidase (SA-ß-gal), an activation marker of the cellular senescence program, as well as the production of cytokines and chemokines involved with the senescence-associated secretory phenotype (SASP) such as IL-6, TNF-α, IL-1ß, and MCP-1. Infected fibroblasts released increased amounts of stress-associated factors nitric oxide (NO) and reactive oxygen species (ROS), and the treatment with antioxidants deferoxamine (DFO) and N-acetylcysteine reduced ROS generation, secretion of SASP-related cytokine IL-6, SA-ß-gal activity, and parasite load by infected fibroblasts. Taken together, our data suggest that T. cruzi infection triggers a rapid cellular stress response followed by induction of a senescent-like phenotype in NIH-3T3 fibroblasts, enabling them to act as reservoirs of parasites during the early stages of the Chagas disease.

6.
Front Immunol ; 9: 671, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29706955

RESUMEN

Few studies investigate the major protein antigens targeted by the antibody diversity of infected mice with Trypanosoma cruzi. To detect global IgG antibody specificities, sera from infected mice were immunoblotted against whole T. cruzi extracts. By proteomic analysis, we were able to identify the most immunogenic T. cruzi proteins. We identified three major antigens as pyruvate phosphate dikinase, Hsp-85, and ß-tubulin. The major protein band recognized by host IgG was T. cruzi ß-tubulin. The T. cruzi ß-tubulin gene was cloned, expressed in E. coli, and recombinant T. cruzi ß-tubulin was obtained. Infection increased IgG reactivity against recombinant T. cruzi ß-tubulin. A single immunization of mice with recombinant T. cruzi ß-tubulin increased specific IgG reactivity and induced protection against T. cruzi infection. These results indicate that repertoire analysis is a valid approach to identify antigens for vaccines against Chagas disease.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Enfermedad de Chagas/inmunología , Inmunoglobulina G/inmunología , Proteínas Protozoarias/inmunología , Trypanosoma cruzi/inmunología , Tubulina (Proteína)/inmunología , Animales , Modelos Animales de Enfermedad , Inmunización , Masculino , Ratones Endogámicos BALB C , Ratones Mutantes
7.
Front Immunol ; 8: 1560, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29204144

RESUMEN

As key cells, able to host and kill Leishmania parasites, inflammatory monocytes/macrophages are potential vaccine and therapeutic targets to improve immune responses in Leishmaniasis. Macrophage phenotypes range from M1, which express NO-mediated microbial killing, to M2 macrophages that might help infection. Resistance to Leishmaniasis depends on Leishmania species, mouse strain, and both innate and adaptive immunity. C57BL/6 (B6) mice are resistant and control infection, whereas Leishmania parasites thrive in BALB/c mice, which are susceptible to develop cutaneous lesions in the course of infection with Leishmania major, but not upon infection with Leishmania braziliensis. Here, we investigated whether a deficit in early maturation of inflammatory monocytes into macrophages in BALB/c mice underlies increased susceptibility to L. major versus L. braziliensis parasites. We show that, after infection with L. braziliensis, monocytes are recruited to peritoneum, differentiate into macrophages, and develop an M1 phenotype able to produce proinflammatory cytokines in both B6 and BALB/c mice. Nonetheless, more mature macrophages from B6 mice expressed inducible NO synthase (iNOS) and higher NO production in response to L. braziliensis parasites, whereas BALB/c mice developed macrophages expressing an incomplete M1 phenotype. By contrast, monocytes recruited upon L. major infection gave rise to immature macrophages that failed to induce an M1 response in BALB/c mice. Overall, these results are consistent with the idea that resistance to Leishmania infection correlates with improved maturation of macrophages in a mouse-strain and Leishmania-species dependent manner. All-trans retinoic acid (ATRA) has been proposed as a therapy to differentiate immature myeloid cells into macrophages and help immunity to tumors. To prompt monocyte to macrophage maturation upon L. major infection, we treated B6 and BALB/c mice with ATRA. Unexpectedly, treatment with ATRA reduced proinflammatory cytokines, iNOS expression, and parasite killing by macrophages. Moreover, ATRA promoted an M1 to M2 transition in bone marrow-derived macrophages from both strains. Therefore, ATRA uncouples macrophage maturation and development of M1 phenotype and downmodulates macrophage-mediated immunity to L. major parasites. Cautions should be taken for the therapeutic use of ATRA, by considering direct effects on innate immunity to intracellular pathogens.

8.
PLoS One ; 9(1): e85715, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24416445

RESUMEN

We investigated early cellular responses induced by infection with Leishmania major in macrophages from resistant C57/BL6 mice. Infection increased production of reactive oxygen species by resident, but not inflammatory peritoneal macrophages. In addition, infection increased activation of stress-activated protein kinases/c-Jun N-terminal kinases (SAPK/JNK) in resident, but not in inflammatory peritoneal macrophages. Infection also increased expression of membrane and soluble FasL, but infected macrophages remained viable after 48 h. Infection increased secretion of cytokines/chemokines TNF-α, IL-6, TIMP-1, IL-1RA, G-CSF, TREM, KC, MIP-1α, MIP-1ß, MCP-1, and MIP-2 in resident macrophages. Addition of antioxidants deferoxamine and N-acetylcysteine reduced ROS generation and JNK activation. Addition of antioxidants or JNK inhibitor SP600125 reduced secretion of KC. Furthermore, treatment with antioxidants or JNK inhibitor also reduced intracellular parasite replication. These results indicated that infection triggers a rapid cellular stress response in resident macrophages which induces proinflammatory signals, but is also involved in parasite survival and replication in host macrophages.


Asunto(s)
Leishmania major/fisiología , Leishmaniasis Cutánea/patología , Leishmaniasis Cutánea/parasitología , Macrófagos/patología , Macrófagos/parasitología , Estrés Fisiológico , Animales , Antioxidantes/metabolismo , Muerte Celular/efectos de los fármacos , Quimiocinas/biosíntesis , Proteína Ligando Fas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Leishmania major/efectos de los fármacos , Leishmania major/crecimiento & desarrollo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Parásitos/efectos de los fármacos , Parásitos/crecimiento & desarrollo , Parásitos/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Estrés Fisiológico/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
9.
J Leukoc Biol ; 95(2): 347-55, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24072877

RESUMEN

We investigated how apoptosis pathways mediated by death receptors and caspase-8 affect cytokine responses and immunity to Leishmania major parasites. Splenic CD4 T cells undergo activation-induced apoptosis, and blockade of FasL-Fas interaction increased IFN-γ and IL-4 cytokine responses to L. major antigens. To block death receptor-induced death, we used mice expressing a T cell-restricted transgene for vFLIP. Inhibition of caspase-8 activation in vFLIP mice enhanced Th1 and Th2 cytokine responses to L. major infection, even in the Th1-prone B6 background. We also observed increased NO production by splenocytes from vFLIP mice upon T cell activation. Despite an exacerbated Th2 response, vFLIP mice controlled better L. major infection, with reduced lesions and lower parasite loads compared with WT mice. Moreover, injection of anti-IL-4 mAb in infected vFLIP mice disrupted control of parasite infection. Therefore, blockade of caspase-8 activity in T cells improves immunity to L. major infection by promoting increased Th1 and Th2 responses.


Asunto(s)
Caspasa 8/metabolismo , Inmunidad Celular/inmunología , Leishmania major/inmunología , Leishmaniasis/inmunología , Leishmaniasis/prevención & control , Células TH1/inmunología , Células Th2/inmunología , Animales , Antígenos de Protozoos/inmunología , Apoptosis , Femenino , Humanos , Interleucina-4/metabolismo , Leishmaniasis/parasitología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Virales/inmunología
10.
J Parasitol Res ; 2012: 250793, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22619699
11.
PLoS Pathog ; 8(5): e1002699, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615561

RESUMEN

MHC class Ia-restricted CD8(+) T cells are important mediators of the adaptive immune response against infections caused by intracellular microorganisms. Whereas antigen-specific effector CD8(+) T cells can clear infection caused by intracellular pathogens, in some circumstances, the immune response is suboptimal and the microorganisms survive, causing host death or chronic infection. Here, we explored the cellular and molecular mechanisms that could explain why CD8(+) T cell-mediated immunity during infection with the human protozoan parasite Trypanosoma cruzi is not optimal. For that purpose, we compared the CD8(+) T-cell mediated immune responses in mice infected with T. cruzi or vaccinated with a recombinant adenovirus expressing an immunodominant parasite antigen. Several functional and phenotypic characteristics of specific CD8(+) T cells overlapped. Among few exceptions was an accelerated expansion of the immune response in adenoviral vaccinated mice when compared to infected ones. Also, there was an upregulated expression of the apoptotic-signaling receptor CD95 on the surface of specific T cells from infected mice, which was not observed in the case of adenoviral-vaccinated mice. Most importantly, adenoviral vaccine provided at the time of infection significantly reduced the upregulation of CD95 expression and the proapoptotic phenotype of pathogen-specific CD8(+) cells expanded during infection. In parallel, infected adenovirus-vaccinated mice had a stronger CD8 T-cell mediated immune response and survived an otherwise lethal infection. We concluded that a suboptimal CD8(+) T-cell response is associated with an upregulation of CD95 expression and a proapoptotic phenotype. Both can be blocked by adenoviral vaccination.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neuraminidasa/inmunología , Vacunas Antiprotozoos/inmunología , Trypanosoma cruzi/inmunología , Receptor fas/biosíntesis , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Apoptosis , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/prevención & control , Interferón gamma/biosíntesis , Proteína 1 de la Membrana Asociada a los Lisosomas/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trypanosoma cruzi/patogenicidad , Vacunas Sintéticas/inmunología
12.
J Leukoc Biol ; 90(6): 1191-7, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21934068

RESUMEN

Th1/Th2 cytokines play a key role in immune responses to Leishmania major by controlling macrophage activation for NO production and parasite killing. MDSCs, including myeloid precursors and immature monocytes, produce NO and suppress T cell responses in tumor immunity. We hypothesized that NO-producing MDSCs could help immunity to L. major infection. Gr1(hi)(Ly6C(hi)) CD11b(hi) MDSCs elicited by L. major infection suppressed polyclonal and antigen-specific T cell proliferation. Moreover, L. major-induced MDSCs killed intracellular parasites in a NO-dependent manner and reduced parasite burden in vivo. By contrast, treatment with ATRA, which induces MDSCs to differentiate into macrophages, increased development of lesions, parasite load, and T cell proliferation in draining LNs. Altogether, these results indicate that NO-producing MDSCs help protective immunity to L. major infection, despite suppressed T cell proliferation.


Asunto(s)
Inmunidad Celular , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Células Mieloides/inmunología , Células Madre/inmunología , Linfocitos T/inmunología , Animales , Células Cultivadas , Resistencia a la Enfermedad/inmunología , Terapia de Inmunosupresión , Leishmaniasis Cutánea/parasitología , Leishmaniasis Cutánea/patología , Masculino , Ratones , Ratones Endogámicos , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/parasitología , Células Mieloides/metabolismo , Células Mieloides/parasitología , Células Madre/parasitología , Células Madre/patología , Linfocitos T/metabolismo , Linfocitos T/parasitología
13.
Eur J Immunol ; 40(2): 417-25, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19950177

RESUMEN

Phagocytic removal of apoptotic lymphocytes exacerbates replication of Trypanosoma cruzi in macrophages. We investigated the presence of Ab against apoptotic lymphocytes in T. cruzi infection and the role of these Ab in parasite replication. Both control and chagasic serum contained IgG Ab that opsonized apoptotic lymphocytes. Treatment of apoptotic lymphocytes with purified IgG from chagasic, but not control serum, reduced T. cruzi replication in macrophages. The protective effect of chagasic IgG depended on Fcgamma receptors, as demonstrated by the requirement for the intact Fc portion of IgG, and the effect could be abrogated by treating macrophages with an anti-CD16/CD32 Fab fragment. Chagasic IgG displayed increased reactivity against a subset of apoptotic cell Ag, as measured by flow cytometry and immunoblot analyses. Apoptotic lymphocytes treated with chagasic IgG, but not control IgG, increased production of TNF-alpha, while decreasing production of TGF-beta1 by infected macrophages. Increased control of parasite replication required TNF-alpha production. Previous immunization with apoptotic cells or injection of apoptotic cells opsonized with chagasic IgG reduced parasitemia in infected mice. These results indicate that Ab raised against apoptotic cells could play a protective role in control of T. cruzi replication by macrophages.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Enfermedad de Chagas/inmunología , Linfocitos/inmunología , Macrófagos/inmunología , Trypanosoma cruzi/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Traslado Adoptivo , Animales , Anticuerpos Antiprotozoarios/farmacología , Apoptosis , Células Cultivadas , Enfermedad de Chagas/parasitología , Enfermedad de Chagas/terapia , Técnicas de Cocultivo , Citometría de Flujo , Immunoblotting , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Linfocitos/citología , Linfocitos/efectos de los fármacos , Macrófagos/citología , Macrófagos/parasitología , Masculino , Ratones , Ratones Endogámicos BALB C , Parasitemia/inmunología , Parasitemia/parasitología , Parasitemia/terapia , Fagocitosis , Factor de Crecimiento Transformador beta1/metabolismo , Trypanosoma cruzi/efectos de los fármacos , Trypanosoma cruzi/crecimiento & desarrollo
14.
Mem Inst Oswaldo Cruz ; 104 Suppl 1: 259-62, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19753482

RESUMEN

Host cell apoptosis plays an important immune regulatory role in parasitic infections. Infection of mice with Trypanosoma cruzi, the causative agent of Chagas disease, induces lymphocyte apoptosis. In addition, phagocytosis of apoptotic cells stimulates the growth of T. cruzi inside host macrophages. In spite of progress made in this area, the importance of apoptosis in the pathogenesis of Chagas disease remains unclear. Here we review the evidence of apoptosis in mice and humans infected with T. cruzi. We also discuss the mechanisms by which apoptosis can influence underlying host responses and tissue damage during Chagas disease progression.


Asunto(s)
Apoptosis/inmunología , Enfermedad de Chagas/inmunología , Interacciones Huésped-Parásitos/inmunología , Trypanosoma cruzi/fisiología , Animales , Enfermedad de Chagas/parasitología , Enfermedad de Chagas/patología , Progresión de la Enfermedad , Humanos , Inmunidad Celular , Ratones , Fagocitosis/inmunología , Trypanosoma cruzi/inmunología
15.
Mem. Inst. Oswaldo Cruz ; 104(supl.1): 259-262, July 2009.
Artículo en Inglés | LILACS | ID: lil-520887

RESUMEN

Host cell apoptosis plays an important immune regulatory role in parasitic infections. Infection of mice with Trypanosoma cruzi, the causative agent of Chagas disease, induces lymphocyte apoptosis. In addition, phagocytosis of apoptotic cells stimulates the growth of T. cruzi inside host macrophages. In spite of progress made in this area, the importance of apoptosis in the pathogenesis of Chagas disease remains unclear. Here we review the evidence of apoptosis in mice and humans infected with T. cruzi. We also discuss the mechanisms by which apoptosis can influence underlying host responses and tissue damage during Chagas disease progression.


Asunto(s)
Animales , Humanos , Ratones , Apoptosis/inmunología , Enfermedad de Chagas/inmunología , Interacciones Huésped-Parásitos/inmunología , Trypanosoma cruzi/fisiología , Enfermedad de Chagas/parasitología , Enfermedad de Chagas/patología , Progresión de la Enfermedad , Inmunidad Celular , Fagocitosis/inmunología , Trypanosoma cruzi/inmunología
16.
Immunopharmacol Immunotoxicol ; 31(2): 159-73, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18785049

RESUMEN

Caspases are cysteine aspartases acting either as initiators (caspases 8, 9, and 10) or executioners (caspases 3, 6, and 7) to induce programmed cell death by apoptosis. Parasite infections by certain intracellular protozoans increase host cell life span by targeting caspase activation. Conversely, caspase activation, followed by apoptosis of lymphocytes and other cells, prevents effective immune responses to chronic parasite infection. Here we discuss how pharmacological inhibition of caspases might affect the immunity to protozoan infections, by either blocking or delaying apoptosis.


Asunto(s)
Antiprotozoarios/uso terapéutico , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Infecciones por Protozoos/tratamiento farmacológico , Animales , Antiprotozoarios/inmunología , Apoptosis/inmunología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Ratones , Infecciones por Protozoos/enzimología , Infecciones por Protozoos/inmunología , Receptores de Muerte Celular/inmunología
17.
An Acad Bras Cienc ; 80(1): 129-36, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18345381

RESUMEN

Following infection with Leishmania major, T cell activation and apoptosis can be detected in draining lymph nodes of C57BL/6-infected mice. We investigated the mechanisms involved in apoptosis and cytokine expression following T cell activation. After two weeks of infection, apoptotic T cells were not detected in draining lymph nodes but activation with anti-CD3 induced apoptosis in both CD4 and CD8 T cells. Treatment with anti-Fas Ligand, caspase-8 or caspase- 9 inhibitors did not block activation-induced T-cell death. We also investigated whether the blockade of caspase-8 activity would affect the expression of type-1 or type-2 cytokines. At early stages of infection, both CD4 and CD8 T cells expressed IFN-gamma upon activation. Treatment with the caspase-8 inhibitor zIETD-fmk (benzyl-oxycarbonyl-Ile- Glu(OMe)-Thr-Asp(OMe)-fluoromethyl ketone) reduced the proportion of CD8 T cells and IFN-gamma expression in both CD4 and CD8 T cells. We conclude that a non apoptotic role of caspase-8 activity may be required for T cell-mediated type-1 responses during L. major infection.


Asunto(s)
Apoptosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inhibidores de Caspasas , Interferón gamma/inmunología , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD8-positivos/enzimología , Inhibidores de Cisteína Proteinasa/farmacología , Femenino , Inmunidad Celular , Leishmaniasis Cutánea/parasitología , Ganglios Linfáticos/parasitología , Ratones , Ratones Endogámicos C57BL
18.
An. acad. bras. ciênc ; 80(1): 129-136, Mar. 2008. graf
Artículo en Inglés | LILACS | ID: lil-477420

RESUMEN

Following infection with Leishmania major, T cell activation and apoptosis can be detected in draining lymph nodes of C57BL/6-infected mice. We investigated the mechanisms involved in apoptosis and cytokine expression following Tcellactivation. After two weeks of infection, apoptotic T cells were not detected in draining lymph nodes but activation with anti-CD3 induced apoptosis in both CD4 and CD8 T cells. Treatment with anti-FasLigand, caspase-8 or caspase- 9 inhibitors did not block activation-induced T-cell death. We also investigated whether the blockade of caspase-8 activity would affect the expression of type-1 or type-2 cytokines. At early stages of infection, both CD4 and CD8 T cells expressed IFN-gamma upon activation. Treatment with the caspase-8 inhibitor zIETD-fmk (benzyl-oxycarbonyl-Ile- Glu(OMe)-Thr-Asp(OMe)-fluoromethyl ketone) reduced the proportion of CD8 T cells and IFN-gamma expression in both CD4 and CD8T cells. We conclude that a non apoptotic role of caspase-8 activity may be required for T cell-mediated type-1 responses during L. major infection.


A ativação e a morte por apoptose de linfócitos T foram observadas em linfonodos drenantes de camundongos C57BL/6 infectados com Leishmania major. Investigamos os mecanismos envolvidos na apoptose e na expressão de citocinas após a ativação de linfócitos T. Após duas semanas de infecção, embora as células apoptóticas ainda não sejam detectadas em linfonodos drenantes, células T CD4 e CD8 sofrem apoptose após ativação com anti-CD3. O tratamento com anticorpo antagonista anti-Ligante de Fas, ou com inibidores das caspases-8 e 9, não bloqueou a morte induzida por ativação das células T. Investigamos também se a inibição da atividade da caspase-8 poderia afetar a expressão de citocinas tipo-1 ou tipo-2. Nos estágios iniciais da infecção, células T CD4 e CD8 de animais infectados com L. major expressaram IFN-gama após ativação. O tratamento com o inibidor de caspase-8 zIETD (benzoil-oxicarbonil-Ile-Glu(OMe)-Thr-Asp(OMe)-fluorometilcetona) durante a estimulação de células T reduziu a proporção de células T CD8 e a expressão de IFN-gama por células T CD4 e CD8. Concluimos que a atividade não apoptótica de caspase-8 pode ser necessária para o estabelecimento da imunidade mediada por células T durante a infecção por L. major.


Asunto(s)
Animales , Femenino , Ratones , Apoptosis/inmunología , /inmunología , /inmunología , /antagonistas & inhibidores , Interferón gamma/inmunología , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Clorometilcetonas de Aminoácidos/farmacología , /enzimología , /enzimología , Inhibidores de Cisteína Proteinasa/farmacología , Inmunidad Celular , Leishmaniasis Cutánea/parasitología , Ganglios Linfáticos/parasitología
19.
Eur J Immunol ; 38(1): 139-46, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18085669

RESUMEN

Infection with Trypanosoma cruzi causes expansion of subcutaneous (SLN) and atrophy of mesenteric (MLN) lymph nodes. Here we show that excision of MLN increased parasitemia in T. cruzi-infected mice. We then studied how apoptosis of MLN cells affects immune responses to infection. T cell apoptosis increased in the MLN compared to SLN in T. cruzi-infected mice. Absolute numbers of naïve T cells decreased, and activated T cells failed to accumulate in MLN during infection. In addition, activated T cells from MLN produced less IL-2, IFN-gamma, IL-4, and IL-10 than T cells from SLN. Treatment with IL-4 or with caspase-9 inhibitor increased the recovery of viable T cells in vitro. Treatment with caspase-9 inhibitor also increased the production of cytokines by MLN T cells from infected mice. Moreover, injection of a pan caspase inhibitor prevented MLN atrophy during T. cruzi infection. Caspase-9, but not caspase-8, inhibitor also reduced MLN atrophy and increased the recovery of naïve and activated T cells from MLN. These findings indicate that caspase-mediated apoptosis and defective cytokine production are implicated in MLN atrophy and affect immune responses to T. cruzi infection.


Asunto(s)
Apoptosis/inmunología , Enfermedad de Chagas/inmunología , Ganglios Linfáticos/inmunología , Mesenterio/inmunología , Piel/inmunología , Linfocitos T/inmunología , Animales , Atrofia , Caspasas/efectos de los fármacos , Caspasas/inmunología , Caspasas/metabolismo , Citocinas/biosíntesis , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/patología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Linfocitos T/microbiología , Linfocitos T/patología , Trypanosoma cruzi
20.
J Immunol ; 179(6): 3988-94, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17785837

RESUMEN

We investigated the role of neutrophil elastase (NE) in interactions between murine inflammatory neutrophils and macrophages infected with the parasite Leishmania major. A blocker peptide specific for NE prevented the neutrophils from inducing microbicidal activity in macrophages. Inflammatory neutrophils from mutant pallid mice were defective in the spontaneous release of NE, failed to induce microbicidal activity in wild-type macrophages, and failed to reduce parasite loads upon transfer in vivo. Conversely, purified NE activated macrophages and induced microbicidal activity dependent on secretion of TNF-alpha. Induction of macrophage microbicidal activity by either neutrophils or purified NE required TLR4 expression by macrophages. Injection of purified NE shortly after infection in vivo reduced the burden of L. major in draining lymph nodes of TLR4-sufficient, but not TLR4-deficient mice. These results indicate that NE plays a previously unrecognized protective role in host responses to L. major infection.


Asunto(s)
Líquido Intracelular/inmunología , Líquido Intracelular/parasitología , Leishmania major/inmunología , Elastasa de Leucocito/fisiología , Macrófagos/inmunología , Macrófagos/parasitología , Neutrófilos/inmunología , Receptor Toll-Like 4/metabolismo , Traslado Adoptivo , Animales , Células Cultivadas , Técnicas de Cocultivo , Activación Enzimática/inmunología , Humanos , Líquido Intracelular/enzimología , Leishmania major/crecimiento & desarrollo , Elastasa de Leucocito/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/enzimología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Neutrófilos/enzimología , Neutrófilos/patología , Neutrófilos/trasplante , Transporte de Proteínas/inmunología , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA