Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 17(4): 356-63, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27002843

RESUMEN

Innate cells are responsible for the rapid recognition of infection and mediate essential mechanisms of pathogen elimination, and also facilitate adaptive immune responses. We review here the numerous intricate interactions among innate cells that initiate protective immunity. The efficient eradication of pathogens depends on the coordinated actions of multiple cells, including innate cells and epithelial cells. Rather than acting as isolated effector cells, innate cells are in constant communication with other responding cells of the immune system, locally and distally. These interactions are critically important for the efficient control of primary infections as well for the development of 'trained' innate cells that facilitate the rapid elimination of homologous or heterologous infections.


Asunto(s)
Inmunidad Adaptativa/inmunología , Citocinas/inmunología , Inmunidad Innata/inmunología , Infecciones/inmunología , Células Asesinas Naturales/inmunología , Células Mieloides/inmunología , Animales , Basófilos/inmunología , Eosinófilos/inmunología , Humanos , Macrófagos/inmunología , Mastocitos/inmunología , Monocitos/inmunología , Neutrófilos/inmunología
2.
Nat Immunol ; 15(10): 938-46, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25173346

RESUMEN

We examined the role of innate cells in acquired resistance to the natural murine parasitic nematode, Nippostrongylus brasiliensis. Macrophages obtained from lungs as late as 45 d after N. brasiliensis inoculation were able to transfer accelerated parasite clearance to naive recipients. Primed macrophages adhered to larvae in vitro and triggered increased mortality of parasites. Neutrophil depletion in primed mice abrogated the protective effects of transferred macrophages and inhibited their in vitro binding to larvae. Neutrophils in parasite-infected mice showed a distinct transcriptional profile and promoted alternatively activated M2 macrophage polarization through secretory factors including IL-13. Differentially activated neutrophils in the context of a type 2 immune response therefore prime a long-lived effector macrophage phenotype that directly mediates rapid nematode damage and clearance.


Asunto(s)
Inmunidad Adaptativa/inmunología , Macrófagos/inmunología , Neutrófilos/inmunología , Nippostrongylus/inmunología , Infecciones por Strongylida/inmunología , Animales , Adhesión Celular/inmunología , Adhesión Celular/fisiología , Células Cultivadas , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Resistencia a la Enfermedad/inmunología , Femenino , Citometría de Flujo , Interacciones Huésped-Parásitos/inmunología , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-13/metabolismo , Subunidad alfa del Receptor de Interleucina-4/genética , Subunidad alfa del Receptor de Interleucina-4/inmunología , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Larva/inmunología , Larva/fisiología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/parasitología , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Noqueados , Neutrófilos/metabolismo , Nippostrongylus/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Strongylida/genética , Infecciones por Strongylida/parasitología , Transcriptoma/inmunología
3.
Nat Immunol ; 14(11): 1118-1126, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24145791

RESUMEN

Helminth infections are ubiquitous worldwide and can trigger potent immune responses that differ from and potentially antagonize host protective responses to microbial pathogens. In this Review we focus on the three main killers in infectious disease-AIDS, tuberculosis and malaria-and critically assesses whether helminths adversely influence host control of these diseases. We also discuss emerging concepts for how M2 macrophages and helminth-modulated dendritic cells can potentially influence the protective immune response to concurrent infections. Finally, we present evidence advocating for more efforts to determine how and to what extent helminths interfere with the successful control of specific concurrent coinfections.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/inmunología , Células Dendríticas/inmunología , Helmintiasis/inmunología , Macrófagos/inmunología , Malaria/inmunología , Tuberculosis/inmunología , Síndrome de Inmunodeficiencia Adquirida/epidemiología , Síndrome de Inmunodeficiencia Adquirida/virología , África/epidemiología , Animales , Asia/epidemiología , Coinfección , Células Dendríticas/microbiología , Células Dendríticas/parasitología , Células Dendríticas/virología , Helmintiasis/epidemiología , Helmintiasis/parasitología , Helmintos/inmunología , Interacciones Huésped-Parásitos , Humanos , América Latina/epidemiología , Macrófagos/microbiología , Macrófagos/parasitología , Macrófagos/virología , Malaria/epidemiología , Malaria/parasitología , Tuberculosis/epidemiología , Tuberculosis/microbiología
4.
Proc Natl Acad Sci U S A ; 119(35): e2123267119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35994660

RESUMEN

The pregnant uterus is an immunologically rich organ, with dynamic changes in the inflammatory milieu and immune cell function underlying key stages of pregnancy. Recent studies have implicated dysregulated expression of the interleukin-1 (IL-1) family cytokine, IL-33, and its receptor, ST2, in poor pregnancy outcomes in women, including recurrent pregnancy loss, preeclampsia, and preterm labor. How IL-33 supports pregnancy progression in vivo is not well understood. Here, we demonstrate that maternal IL-33 signaling critically regulates uterine tissue remodeling and immune cell function during early pregnancy in mice. IL-33-deficient dams exhibit defects in implantation chamber formation and decidualization, and abnormal vascular remodeling during early pregnancy. These defects coincide with delays in early embryogenesis, increased resorptions, and impaired fetal and placental growth by late pregnancy. At a cellular level, myometrial fibroblasts, and decidual endothelial and stromal cells, are the main IL-33+ cell types in the uterus during decidualization and early placentation, whereas ST2 is expressed by uterine immune populations associated with type 2 immune responses, including ILC2s, Tregs, CD4+ T cells, M2- and cDC2-like myeloid cells, and mast cells. Early pregnancy defects in IL-33-deficient dams are associated with impaired type 2 cytokine responses by uterine lymphocytes and fewer Arginase-1+ macrophages in the uterine microenvironment. Collectively, our data highlight a regulatory network, involving crosstalk between IL-33-producing nonimmune cells and ST2+ immune cells at the maternal-fetal interface, that critically supports pregnancy progression in mice. This work has the potential to advance our understanding of how IL-33 signaling may support optimal pregnancy outcomes in women.


Asunto(s)
Interleucina-33 , Placenta , Placentación , Útero , Animales , Decidua/irrigación sanguínea , Decidua/citología , Decidua/crecimiento & desarrollo , Decidua/inmunología , Femenino , Feto/inmunología , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/deficiencia , Interleucina-33/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Placenta/inmunología , Placenta/metabolismo , Embarazo , Útero/irrigación sanguínea , Útero/crecimiento & desarrollo , Útero/inmunología , Útero/metabolismo
5.
Trends Immunol ; 42(2): 151-164, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33386241

RESUMEN

Type 2 immune responses are typically associated with protection against helminth infections and also with harmful inflammation in response to allergens. Recent advances have revealed that type 2 immunity also contributes to sterile inflammation, cancer, and microbial infections. However, the early events that initiate type 2 immune responses remain poorly defined. New insights reveal major contributions from danger-associated molecular patterns (DAMPs) in the initiation of type 2 immune responses. In this review, we examine the molecules released by the host and pathogens and the role they play in mediating the initiation of mammalian innate type 2 immune responses under a variety of conditions.


Asunto(s)
Helmintiasis , Inmunidad Innata , Alarminas , Alérgenos , Animales , Humanos , Inflamación
6.
Immunity ; 42(2): 216-226, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25692699

RESUMEN

Type 2 inflammatory responses can be elicited by diverse stimuli, including toxins, venoms, allergens, and infectious agents, and play critical roles in resistance and tolerance associated with infection, wound healing, tissue repair, and tumor development. Emerging data suggest that in addition to characteristic type 2-associated cytokines, the epidermal growth factor (EGF)-like molecule Amphiregulin (AREG) might be a critical component of type 2-mediated resistance and tolerance. Notably, numerous studies demonstrate that in addition to the established role of epithelial- and mesenchymal-derived AREG, multiple leukocyte populations including mast cells, basophils, group 2 innate lymphoid cells (ILC2s), and a subset of tissue-resident regulatory CD4(+) T cells can express AREG. In this review, we discuss recent advances in our understanding of the AREG-EGF receptor pathway and its involvement in infection and inflammation and propose a model for the function of this pathway in the context of resistance and tissue tolerance.


Asunto(s)
Familia de Proteínas EGF/inmunología , Receptores ErbB/inmunología , Tolerancia Inmunológica/inmunología , Inflamación/inmunología , Cicatrización de Heridas/inmunología , Anfirregulina , Animales , Helmintiasis/inmunología , Humanos , Gripe Humana/inmunología , Ratones , Neoplasias/inmunología , Infecciones por Orthomyxoviridae/inmunología , Regeneración , Escape del Tumor/inmunología
7.
J Immunol ; 209(11): 2160-2171, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36426972

RESUMEN

More than 2 billion people worldwide are infected with helminths. Thus, it is possible for individuals to experience concomitant infection with helminth and intracellular microbes. Although the helminth-induced type 2 response can suppress type 1 proinflammatory responses required for the immunity against intracellular pathogens in the context of a coinfection, conflicting evidence suggest that helminth infection can enhance antimicrobial immunity. Using a coinfection model with the intestinal helminth Heligmosomoides polygyrus followed by infection with Toxoplasma gondii in Mus Musculus, we showed that the complex and dynamic effect of helminth infection is highly suppressive during the innate phase (days 0-3) of T. gondii infection and less stringent during the acute phase (d10). Helminth coinfection had a strong suppressive effect on the neutrophil, monocytic, and early IFN-γ/IL-12 responses. The IFN-γ response was later restored by compensatory production from T cells despite decreased effector differentiation of T. gondii-specific CD8 T cells. In accordance with the attenuated IFN-γ response, parasite loads were elevated during the acute phase (d10) of T. gondii infection but were transiently controlled by the compensatory T cell response. Unexpectedly, 40% of helminth-coinfected mice exhibited a sustained weight loss phenotype during the postacute phase (d14-18) that was not associated with T. gondii outgrowth, indicating that coinfection led to decreased disease tolerance during T. gondii infection. Our work uncovers the dynamic nature of the helminth immunomodulatory effects on concomitant infections or immune responses and unveils a loss of disease tolerance phenotype triggered by coinfection with intestinal helminth.


Asunto(s)
Coinfección , Nematospiroides dubius , Toxoplasma , Toxoplasmosis , Animales , Ratones , Tolerancia Inmunológica
8.
FASEB J ; 35(11): e21935, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34591327

RESUMEN

Inosine monophosphate (IMP) is the intracellular precursor for both adenosine monophosphate and guanosine monophosphate and thus plays a central role in intracellular purine metabolism. IMP can also serve as an extracellular signaling molecule, and can regulate diverse processes such as taste sensation, neutrophil function, and ischemia-reperfusion injury. How IMP regulates inflammation induced by bacterial products or bacteria is unknown. In this study, we demonstrate that IMP suppressed tumor necrosis factor (TNF)-α production and augmented IL-10 production in endotoxemic mice. IMP exerted its effects through metabolism to inosine, as IMP only suppressed TNF-α following its CD73-mediated degradation to inosine in lipopolysaccharide-activated macrophages. Studies with gene targeted mice and pharmacological antagonism indicated that A2A , A2B, and A3 adenosine receptors are not required for the inosine suppression of TNF-α production. The inosine suppression of TNF-α production did not require its metabolism to hypoxanthine through purine nucleoside phosphorylase or its uptake into cells through concentrative nucleoside transporters indicating a role for alternative metabolic/uptake pathways. Inosine augmented IL-ß production by macrophages in which inflammasome was activated by lipopolysaccharide and ATP. In contrast to its effects in endotoxemia, IMP failed to affect the inflammatory response to abdominal sepsis and pneumonia. We conclude that extracellular IMP and inosine differentially regulate the inflammatory response.


Asunto(s)
Endotoxemia/metabolismo , Inosina Monofosfato/metabolismo , Inosina/metabolismo , Neumonía Neumocócica/metabolismo , Streptococcus pneumoniae , Antagonistas del Receptor de Adenosina A2/farmacología , Antagonistas del Receptor de Adenosina A3/farmacología , Animales , Modelos Animales de Enfermedad , Interleucina-10/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Neumonía Neumocócica/microbiología , Quinazolinas/farmacología , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2B/metabolismo , Receptor de Adenosina A3/metabolismo , Transducción de Señal/efectos de los fármacos , Triazoles/farmacología , Factor de Necrosis Tumoral alfa/biosíntesis
9.
Purinergic Signal ; 18(3): 345-358, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35838900

RESUMEN

Extracellular adenosine is a biologically active signaling molecule that accumulates at sites of metabolic stress in sepsis. Extracellular adenosine has potent immunosuppressive effects by binding to and activating G protein-coupled A2A adenosine receptors (A2AARs) on the surface of neutrophils. A2AAR signaling reproduces many of the phenotypic changes in neutrophils that are characteristic of sepsis, including decreased degranulation, impaired chemotaxis, and diminished ability to ingest and kill bacteria. We hypothesized that A2AARs also suppress neutrophil aging, which precedes cell death, and N1 to N2 polarization. Using human neutrophils isolated from healthy subjects, we demonstrate that A2AAR stimulation slows neutrophil aging, suppresses cell death, and promotes the polarization of neutrophils from an N1 to N2 phenotype. Using genetic knockout and pharmacological blockade, we confirmed that A2AARs decrease neutrophil aging in murine sepsis induced by cecal ligation and puncture. A2AARs expression is increased in neutrophils from septic patients compared to healthy subject but A2AAR expression fails to correlate with aging or N1/N2 polarization. Our data reveals that A2AARs regulate neutrophil aging in healthy but not septic neutrophils.


Asunto(s)
Neutrófilos , Sepsis , Adenosina , Envejecimiento , Animales , Humanos , Ratones , Ratones Noqueados , Neutrófilos/metabolismo , Fenotipo , Receptor de Adenosina A2A/metabolismo
10.
Nat Mater ; 18(3): 289-297, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30664693

RESUMEN

Initiation of the innate sterile inflammatory response that can develop in response to microparticle exposure is little understood. Here, we report that a potent type 2 immune response associated with the accumulation of neutrophils, eosinophils and alternatively activated (M2) macrophages was observed in response to sterile microparticles similar in size to wear debris associated with prosthetic implants. Although elevations in interleukin-33 (IL-33) and type 2 cytokines occurred independently of caspase-1 inflammasome signalling, the response was dependent on Bruton's tyrosine kinase (BTK). IL-33 was produced by macrophages and BTK-dependent expression of IL-33 by macrophages was sufficient to initiate the type 2 response. Analysis of inflammation in patient periprosthetic tissue also revealed type 2 responses under aseptic conditions in patients undergoing revision surgery. These findings indicate that microparticle-induced sterile inflammation is initiated by macrophages activated to produce IL-33. They further suggest that both BTK and IL-33 may provide therapeutic targets for wear debris-induced periprosthetic inflammation.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/metabolismo , Interleucina-33/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Falla de Prótesis , Artroplastia/efectos adversos , Caspasa 1/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-33/biosíntesis , Macrófagos/inmunología , Transducción de Señal/efectos de los fármacos
11.
J Immunol ; 198(2): 634-639, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27956529

RESUMEN

Concurrent helminth infection potently inhibits T cell immunity; however, whether helminthes prevent T cell priming or skew clonal recruitment and effector differentiation is not known. Using coinfection with two natural mouse pathogens, Heligmosomoides polygyrus and Toxoplasma gondii, to investigate the negative impact of helminthes on the CD8 T cell response, we demonstrate helminth-induced suppression of IL-12-dependent differentiation of killer-like receptor G1+ effector CD8 T cells and IFN-γ production. Nevertheless, reversal of helminth suppression of the innate IL-12 response of CD8α+ dendritic cells, which occurred in STAT6-deficient mice, was not sufficient to normalize CD8 T cell differentiation. Instead, a combined deficiency in IL-4 and IL-10 was required to reverse the negative effects of helminth coinfection on the CD8 T cell response. Monoclonal T. gondii-specific CD8 T cells adoptively transferred into coinfected mice recapitulated the spectrum of helminth-induced effects on the polyclonal CD8 T response, indicating the lack of requirement for clonal skewing.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Coinfección/inmunología , Helmintiasis/inmunología , Activación de Linfocitos/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Interleucina-10/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nematospiroides dubius/inmunología , Toxoplasma/inmunología
13.
Infect Immun ; 83(3): 1217-23, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25605766

RESUMEN

Previously we had reported that Nippostrongylus brasiliensis, a helminth with a lung migratory phase, affected host resistance against Mycobacterium tuberculosis infection through the induction of alternatively activated (M2) macrophages. Several helminth species do not have an obligatory lung migratory phase but establish chronic infections in the host that include potent immune downregulatory effects, in part mediated through induction of a FoxP3(+) T regulatory cell (Treg) response. Treg cells exhibit duality in their functions in host defense against M. tuberculosis infection since their depletion leads to enhanced priming of T cells in the lymph nodes and attendant improved control of M. tuberculosis infection, while their presence in the lung granuloma protects against excessive inflammation. Heligmosomoides polygyrus is a strictly murine enteric nematode that induces a strong FoxP3 Treg response in the host. Therefore, in this study we investigated whether host immunity to M. tuberculosis infection would be modulated in mice with chronic H. polygyrus infection. We report that neither primary nor memory immunity conferred by Mycobacterium bovis BCG vaccination was affected in mice with chronic enteric helminth infection, despite a systemic increase in FoxP3(+) T regulatory cells. The findings indicate that anti-M. tuberculosis immunity is not similarly affected by all helminth species and highlight the need to consider this inequality in human coinfection studies.


Asunto(s)
Vacuna BCG/administración & dosificación , Inmunidad Innata , Memoria Inmunológica , Infecciones por Mycobacterium/prevención & control , Infecciones por Strongylida/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Enfermedad Crónica , Coinfección , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Expresión Génica , Pulmón/inmunología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Mycobacterium/inmunología , Infecciones por Mycobacterium/patología , Mycobacterium tuberculosis/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/patología , Linfocitos T Reguladores/patología , Vacunación
14.
PLoS Pathog ; 9(11): e1003771, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244174

RESUMEN

Approximately one-third of the world's population suffers from chronic helminth infections with no effective vaccines currently available. Antibodies and alternatively activated macrophages (AAM) form crucial components of protective immunity against challenge infections with intestinal helminths. However, the mechanisms by which antibodies target these large multi-cellular parasites remain obscure. Alternative activation of macrophages during helminth infection has been linked to signaling through the IL-4 receptor alpha chain (IL-4Rα), but the potential effects of antibodies on macrophage differentiation have not been explored. We demonstrate that helminth-specific antibodies induce the rapid trapping of tissue migrating helminth larvae and prevent tissue necrosis following challenge infection with the natural murine parasite Heligmosomoides polygyrus bakeri (Hp). Mice lacking antibodies (JH (-/-)) or activating Fc receptors (FcRγ(-/-)) harbored highly motile larvae, developed extensive tissue damage and accumulated less Arginase-1 expressing macrophages around the larvae. Moreover, Hp-specific antibodies induced FcRγ- and complement-dependent adherence of macrophages to larvae in vitro, resulting in complete larval immobilization. Antibodies together with helminth larvae reprogrammed macrophages to express wound-healing associated genes, including Arginase-1, and the Arginase-1 product L-ornithine directly impaired larval motility. Antibody-induced expression of Arginase-1 in vitro and in vivo occurred independently of IL-4Rα signaling. In summary, we present a novel IL-4Rα-independent mechanism of alternative macrophage activation that is antibody-dependent and which both mediates anti-helminth immunity and prevents tissue disruption caused by migrating larvae.


Asunto(s)
Anticuerpos Antihelmínticos/inmunología , Diferenciación Celular/inmunología , Macrófagos/inmunología , Nematospiroides dubius/inmunología , Receptores de Superficie Celular/inmunología , Infecciones por Strongylida/inmunología , Animales , Anticuerpos Antihelmínticos/genética , Arginasa/genética , Arginasa/inmunología , Diferenciación Celular/genética , Regulación Enzimológica de la Expresión Génica/genética , Regulación Enzimológica de la Expresión Génica/inmunología , Larva , Ratones , Ratones Noqueados , Receptores de Superficie Celular/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Infecciones por Strongylida/genética
15.
Trends Immunol ; 32(2): 80-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21159556

RESUMEN

Similar T helper (Th)2-type immune responses are generated against different helminth parasites, but the mechanisms that initiate Th2 immunity, and the specific immune components that mediate protection against these parasites, can vary greatly. B cells are increasingly recognized as important during the Th2-type immune response to helminths, and B cell activation might be a target for effective vaccine development. Antibody production is a function of B cells during helminth infection and understanding how polyclonal and antigen-specific antibodies contribute should provide important insights into how protective immunity develops. In addition, B cells might also contribute to the host response against helminths through antibody-independent functions including, antigen presentation, as well as regulatory and effector activity. In this review, we examine the role of B cells during Th2-type immune response to these multicellular parasites.


Asunto(s)
Linfocitos B/inmunología , Helmintos/inmunología , Células Th2/inmunología , Animales , Anticuerpos Antihelmínticos/inmunología , Helmintiasis/inmunología , Helmintiasis/parasitología , Humanos , Vacunación
17.
Life Sci Alliance ; 7(11)2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39179288

RESUMEN

Skin wound healing due to full thickness wounds typically results in fibrosis and scarring, where parenchyma tissue is replaced with connective tissue. A major advance in wound healing research would be to instead promote tissue regeneration. Helminth parasites express excretory/secretory (ES) molecules, which can modulate mammalian host responses. One recently discovered ES protein, TGF-ß mimic (TGM), binds the TGF-ß receptor, though likely has other activities. Here, we demonstrate that topical administration of TGM under a Tegaderm bandage enhanced wound healing and tissue regeneration in an in vivo wound biopsy model. Increased restoration of normal tissue structure in the wound beds of TGM-treated mice was observed during mid- to late-stage wound healing. Both accelerated re-epithelialization and hair follicle regeneration were observed. Further analysis showed differential expansion of myeloid populations at different wound healing stages, suggesting recruitment and reprogramming of specific macrophage subsets. This study indicates a role for TGM as a potential therapeutic option for enhanced wound healing.


Asunto(s)
Fibrosis , Proteínas del Helminto , Regeneración , Cicatrización de Heridas , Animales , Ratones , Proteínas del Helminto/metabolismo , Proteínas del Helminto/farmacología , Piel/metabolismo , Piel/lesiones , Ratones Endogámicos C57BL , Folículo Piloso/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Modelos Animales de Enfermedad , Macrófagos/metabolismo , Repitelización , Masculino
18.
FASEB J ; 26(1): 376-86, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21926236

RESUMEN

Adenosine has been implicated in suppressing the proinflammatory responses of classically activated macrophages induced by Th1 cytokines. Alternative macrophage activation is induced by the Th2 cytokines interleukin (IL)-4 and IL-13; however, the role of adenosine in governing alternative macrophage activation is unknown. We show here that adenosine treatment of IL-4- or IL-13-activated macrophages augments the expression of alternative macrophage markers arginase-1, tissue inhibitor of matrix metalloproteinase-1 (TIMP-1), and macrophage galactose-type C-type lectin-1. The stimulatory effect of adenosine required primarily A(2B) receptors because the nonselective adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA) increased both arginase activity (EC(50)=261.8 nM) and TIMP-1 production (EC(50)=80.67 nM), and both pharmacologic and genetic blockade of A(2B) receptors prevented the effect of NECA. A(2A) receptors also contributed to the adenosine augmentation of IL-4-induced TIMP-1 release, as both adenosine and NECA were less efficacious in augmenting TIMP-1 release by A(2A) receptor-deficient than control macrophages. Of the transcription factors known to drive alternative macrophage activation, CCAAT-enhancer-binding protein ß was required, while cAMP response element-binding protein and signal transducer and activator of transcription 6 were dispensable in mediating the effect of adenosine. We propose that adenosine receptor activation suppresses inflammation and promotes tissue restitution, in part, by promoting alternative macrophage activation.


Asunto(s)
Adenosina/metabolismo , Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2B/metabolismo , Adenosina/farmacología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Animales , Arginasa/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Espacio Extracelular/metabolismo , Inflamación/inmunología , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/inmunología , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/inmunología , Factor de Transcripción STAT6/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , Vasodilatadores/farmacología
19.
J Immunol ; 187(12): 6491-8, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22095717

RESUMEN

Wear debris in joint replacements has been suggested as a cause of associated tissue-damaging inflammation. In this study, we examined whether solid titanium microparticles (mTi) of sufficient size to accumulate as wear debris could stimulate innate or adaptive immunity in vivo. mTi, administered in conjunction with OVA, promoted total and Ag-specific elevations in serum IgE and IgG1. Analysis of transferred transgenic OVA-specific naive T cells further showed that mTi acted as an adjuvant to drive Ag-specific Th2 cell differentiation in vivo. Assessment of the innate response indicated that mTi induced rapid recruitment and differentiation of alternatively activated macrophages in vivo, through IL-4- and TLR4-independent pathways. These studies suggest that solid microparticles alone can act as adjuvants to induce potent innate and adaptive Th2-type immune responses and further suggest that wear debris in joint replacements may have Th2-type inflammatory properties.


Asunto(s)
Tamaño de la Partícula , Transducción de Señal/inmunología , Células Th2/inmunología , Células Th2/metabolismo , Titanio/administración & dosificación , Receptor Toll-Like 4/fisiología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/efectos adversos , Traslado Adoptivo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Femenino , Mediadores de Inflamación/administración & dosificación , Mediadores de Inflamación/efectos adversos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Transducción de Señal/efectos de los fármacos , Células Th2/patología , Titanio/efectos adversos , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
20.
Nat Med ; 12(8): 955-60, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16892038

RESUMEN

Although primary and memory responses against bacteria and viruses have been studied extensively, T helper type 2 (T(H)2) effector mechanisms leading to host protection against helminthic parasites remain elusive. Examination of the intestinal epithelial submucosa of mice after primary and secondary infections by a natural gastrointestinal parasite revealed a distinct immune-cell infiltrate after challenge, featuring interleukin-4-expressing memory CD4(+) T cells that induced IL-4 receptor(hi) (IL-4R(hi)) CD206(+) alternatively activated macrophages. In turn, these alternatively activated macrophages (AAMacs) functioned as important effector cells of the protective memory response contributing to parasite elimination, demonstrating a previously unknown mechanism for host protection against intestinal helminths.


Asunto(s)
Macrófagos/inmunología , Nematodos/inmunología , Infecciones por Nematodos/inmunología , Infecciones por Strongylida/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Arginasa/antagonistas & inhibidores , Ácidos Borónicos/farmacología , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD4-Positivos/trasplante , Complejo IV de Transporte de Electrones/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Interacciones Huésped-Parásitos , Inmunohistoquímica , Inyecciones Intravenosas , Interleucina-4/genética , Larva/enzimología , Larva/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Nematodos/fisiología , Infecciones por Nematodos/parasitología , Factor de Transcripción STAT6/genética , Infecciones por Strongylida/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA