Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunol Lett ; 253: 41-53, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36623708

RESUMEN

The gut comprises the largest body interface with the environment and is continuously exposed to nutrients, food antigens, and commensal microbes, as well as to harmful pathogens. Subsets of both macrophages and dendritic cells (DCs) are present throughout the intestinal tract, where they primarily inhabit the gut-associate lymphoid tissue (GALT), such as Peyer's patches and isolated lymphoid follicles. In addition to their role in taking up and presenting antigens, macrophages and DCs possess extensive functional plasticity and these cells play complementary roles in maintaining immune homeostasis in the gut by preventing aberrant immune responses to harmless antigens and microbes and by promoting host defense against pathogens. The ability of macrophages and DCs to induce either inflammation or tolerance is partially lineage imprinted, but can also be dictated by their activation state, which in turn is determined by their specific microenvironment. These cells express several surface and intracellular receptors that detect danger signals, nutrients, and hormones, which can affect their activation state. DCs and macrophages play a fundamental role in regulating T cells and their effector functions. Thus, modulation of intestinal mucosa immunity by targeting antigen presenting cells can provide a promising approach for controlling pathological inflammation. In this review, we provide an overview on the characteristics, functions, and origins of intestinal macrophages and DCs, highlighting the intestinal microenvironmental factors that influence their functions during homeostasis. Unraveling the mechanisms by which macrophages and DCs regulate intestinal immunity will deepen our understanding on how the immune system integrates endogenous and exogenous signals in order to maintain the host's homeostasis.


Asunto(s)
Tejido Linfoide , Macrófagos , Humanos , Inflamación/metabolismo , Alérgenos/metabolismo , Células Dendríticas , Mucosa Intestinal
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.
Trop Med Infect Dis ; 7(7)2022 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-35878141

RESUMEN

Chagas disease (CD) is a neglected tropical disease caused by Trypanosoma cruzi infection that, despite being discovered over a century ago, remains a public health problem, mainly in developing countries. Since T. cruzi can infect a wide range of mammalian host cells, parasite-host interactions may be critical to infection outcome. The intense immune stimulation that helps the control of the parasite's replication and dissemination may also be linked with the pathogenesis and symptomatology worsening. Here, we discuss the findings that support the notion that excessive immune system stimulation driven by parasite persistence might elicit a progressive loss and collapse of immune functions. In this context, cellular stress and inflammatory responses elicited by T. cruzi induce fibroblast and other immune cell senescence phenotypes that may compromise the host's capacity to control the magnitude of T. cruzi-induced inflammation, contributing to parasite persistence and CD progression. A better understanding of the steps involved in the induction of this chronic inflammatory status, which disables host defense capacity, providing an extra advantage to the parasite and predisposing infected hosts prematurely to immunosenescence, may provide insights to designing and developing novel therapeutic approaches to prevent and treat Chagas disease.

4.
Immunol Lett ; 248: 109-118, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35843361

RESUMEN

Efferocytosis is imperative to maintain lung homeostasis and control inflammation. Populations of lung macrophages are the main efferocytes in this tissue, responsible for controlling immune responses and avoiding unrestrained inflammation and autoimmunity through the expression of a plethora of receptors that recognize multiple 'eat me' signals on apoptotic cells. Efferocytosis is essentially anti-inflammatory and tolerogenic. However, in some situations, apoptotic cells phagocytosis can elicit inflammatory and immunogenic immune responses. Here, we summarized the current knowledge of the mechanisms of efferocytosis, and how any abnormality in this process may have an important contribution to the lung pathophysiology of many chronic inflammatory lung diseases such as asthma, acute lung injury, chronic obstructive pulmonary disease, and cystic fibrosis. Further, we consider the consequences of the dual role of efferocytosis on the susceptibility or resistance to pulmonary microbial infections. Understanding how efferocytosis works in different contexts will be useful to the development of new and more effective strategies to control the diversity of lung diseases.


Asunto(s)
Apoptosis , Enfermedades Pulmonares , Humanos , Inflamación , Pulmón , Fagocitosis
5.
Cancer Immunol Res ; 8(10): 1262-1272, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32819969

RESUMEN

Diabetes mellitus (DM) significantly increases the risk for cancer and cancer progression. Hyperglycemia is the defining characteristic of DM and tightly correlates with a poor prognosis in patients with cancer. The hexosamine biosynthetic pathway (HBP) is emerging as a pivotal cascade linking high glucose, tumor progression, and impaired immune function. Here we show that enhanced glucose flow through the HBP drives cancer progression and immune evasion by increasing O-GlcNAcylation in tumor-associated macrophages (TAM). Increased O-GlcNAc skewed macrophage polarization to a M2-like phenotype supporting tumor progression. Finally, we found an upregulation of M2 markers on TAMs in DM2 patients with colorectal cancer compared with nondiabetic normoglycemic patients. Our results provide evidence for a new and targetable mechanism of cancer immune evasion in patients with hyperglycemia, advocating for strict control of hyperglycemia in patients with cancer.


Asunto(s)
Hiperglucemia/fisiopatología , Evasión Inmune/inmunología , Macrófagos/metabolismo , Animales , Modelos Animales de Enfermedad , Glicosilación , Humanos , Masculino , Ratones , Ratones SCID
6.
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
7.
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.

8.
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.

9.
PLoS One ; 10(5): e0124888, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25933287

RESUMEN

B-1 cells can be differentiated from B-2 cells because they are predominantly located in the peritoneal and pleural cavities and have distinct phenotypic patterns and activation properties. A mononuclear phagocyte derived from B-1 cells (B-1CDP) has been described. As the B-1CDP cells migrate to inflammatory/infectious sites and exhibit phagocytic capacity, the microbicidal ability of these cells was investigated using the Leishmania major infection model in vitro. The data obtained in this study demonstrate that B-1CDP cells are more susceptible to infection than peritoneal macrophages, since B-1CDP cells have a higher number of intracellular amastigotes forms and consequently release a larger number of promastigotes. Exacerbated infection by L. major required lipid bodies/PGE2 and IL-10 by B-1CDP cells. Both infection and the production of IL-10 were decreased when PGE2 production was blocked by NSAIDs. The involvement of IL-10 in this mechanism was confirmed, since B-1CDP cells from IL-10 KO mice are more competent to control L. major infection than cells from wild type mice. These findings further characterize the B-1CDP cells as an important mononuclear phagocyte that plays a previously unrecognized role in host responses to L. major infection, most likely via PGE2-driven production of IL-10.


Asunto(s)
Linfocitos B/parasitología , Dinoprostona/metabolismo , Interleucina-10/metabolismo , Leishmania major/fisiología , Leishmaniasis Cutánea/parasitología , Fagocitos/parasitología , Animales , Aspirina/farmacología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Susceptibilidad a Enfermedades , Interleucina-10/biosíntesis , Leishmania major/efectos de los fármacos , Leishmania major/crecimiento & desarrollo , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Gotas Lipídicas/metabolismo , Macrófagos Peritoneales/parasitología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Parasitemia/inmunología , Parasitemia/parasitología , Fagocitos/efectos de los fármacos , Fenotipo , Prostaglandina-Endoperóxido Sintasas/metabolismo
10.
PLoS One ; 9(3): e90582, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24599360

RESUMEN

Neutrophils are involved in the initial steps of most responses to pathogens and are essential components of the innate immune response. Due to the ability to produce and release various soluble mediators, neutrophils may participate in the regulation of the inflammatory response. Little is known about the role of neutrophils during protozoan infections including infection by Trypanosoma cruzi. In the present study we investigated the importance of inflammatory neutrophils on macrophage activation and T. cruzi replication in vitro, in cells obtained from BALB/c mice and C57Bl/6 mice. Co-cultures of BALB/c apoptotic or live neutrophils with infected peritoneal macrophages resulted in increased replication of the parasites and in the production of TGF-ß and PGE2. The treatment with anti-TGF-ß neutralizing antibody and COX inhibitor blocked the parasite replication in vitro. On the other hand, co-cultures of T. cruzi infected macrophages with live neutrophils isolated from C57BL/6 mice resulted in decreased number of trypomastigotes in culture and increased production of TNF-α and NO. The addition of anti-TNF-α neutralizing antibody or elastase inhibitor resulted in the abolishment of macrophage microbicidal effect and increased parasite replication. Addition of elastase to infected macrophages reduced the replication of the parasites, and on the other hand, addition of a selective inhibitor of iNOS increased parasite growth, suggesting the role of NO in this system. Our findings reveal that neutrophils may regulate T. cruzi experimental infection and determine susceptibility and resistance to infection.


Asunto(s)
Elastasa de Leucocito/fisiología , Macrófagos Peritoneales/parasitología , Neutrófilos/enzimología , Trypanosoma cruzi/inmunología , Animales , Apoptosis , Células Cultivadas , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/parasitología , Técnicas de Cocultivo , Citocinas/metabolismo , Dinoprostona/fisiología , Especificidad del Huésped , Interacciones Huésped-Parásitos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Neutrófilos/parasitología , Óxido Nítrico/fisiología
11.
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
12.
Cell Mol Life Sci ; 68(11): 1863-70, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21369708

RESUMEN

Neutrophils and macrophages are phagocytic cells that cooperate during inflammation and tissue repair. Neutrophils undergo apoptosis and are engulfed by macrophages. Engulfment modulates macrophage activation and microbicidal activity. Infection by Leishmania takes place in the context of tissue repair. This article discusses cellular and molecular mechanisms involved in the intimate cooperation of neutrophils and macrophages in Leishmania infection.


Asunto(s)
Leishmaniasis/inmunología , Macrófagos/inmunología , Neutrófilos/inmunología , Animales , Humanos , Inmunidad Activa , Activación de Macrófagos
13.
J Immunol ; 185(4): 2044-50, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20660352

RESUMEN

Clearance of apoptotic exudate neutrophils (efferocytosis) induces either pro- or anti-inflammatory responses in mouse macrophages depending on host genetic background. In this study, we investigated whether neutrophil efferocytosis induces a stable macrophage phenotype that could be recalled by late restimulation with LPS. Bone marrow-derived macrophages previously stimulated by pro- but not anti-inflammatory neutrophil efferocytosis expressed a regulatory/M2b phenotype characterized by low IL-12 and high IL-10 production following restimulation, increased expression of LIGHT/TNF superfamily 14, Th2-biased T cell responses, and permissive replication of Leishmania major. Induction of regulatory/M2b macrophages required neutrophil elastase activity and was partially dependent on TLR4 signaling. These results suggested that macrophage differentiation to a regulatory phenotype plays a role in resolution of inflammation but could contribute to increased humoral Ab responses and parasite persistence in the infected host.


Asunto(s)
Interleucina-10/metabolismo , Interleucina-12/metabolismo , Macrófagos/inmunología , Neutrófilos/inmunología , Fagocitosis/inmunología , Animales , Apoptosis/inmunología , Células Cultivadas , Inflamación/inmunología , Interferón gamma/inmunología , Interferón gamma/farmacología , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Elastasa de Leucocito/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos , Neutrófilos/citología , Óxido Nítrico/metabolismo , Fagocitosis/efectos de los fármacos , Células Th2/inmunología , Células Th2/metabolismo , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA