Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Signal ; 14(702): eabb3966, 2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34582250

RESUMEN

The cytokines interleukin-4 (IL-4) and IL-13 bind to their shared receptor subunit IL-4Rα to direct the alternative activation of macrophages to promote immunosuppression and wound healing. Activated IL-4 and IL-13 receptors recruit the tyrosine phosphatases SHP-1 and SHP-2, which dephosphorylate and inhibit the IL-4Rα subunit. Here, we report that the immunoreceptor SIRPα spatially restricted SHP-2 to promote IL-4 and IL-13 signaling and the alternative activation of macrophages. This effect required the cytoplasmic ITIMs or ITSMs of SIRPα, which underwent tyrosine phosphorylation by Bruton's tyrosine kinase (Btk) that was activated in response to IL-4 and IL-13. This phosphorylation event resulted in the recruitment of SHP-2 to SIRPα and prevented it from binding to and inhibiting IL-4R and IL-13R. Binding of the ligand CD47 to the SIRPα extracellular domain promoted the Btk-mediated phosphorylation of the SIRPα cytoplasmic domain and hence SHP-2 sequestration. Conversely, loss of SIRPα enabled SHP-2 to bind to the γC and IL-13Rα1 subunits of IL-4R and IL-13R, respectively, and dephosphorylate IL-4Rα, dampening its signaling. Impaired wound healing in Sirpα−/− mice with experimental colitis correlated with a deficit of immunosuppressive macrophages in the colon, a condition that was corrected by transfusion of ex vivo­produced, alternatively activated SIRPαhigh macrophages. These studies reveal a previously unappreciated role for SIRPα in promoting IL-4 and IL-13 signaling and reveal the mechanistic basis by which SIRPα enhances the alternative activation of macrophages.


Asunto(s)
Interleucina-13 , Interleucina-4 , Macrófagos , Transducción de Señal
2.
J Immunol ; 207(5): 1419-1427, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34348974

RESUMEN

Macrophage functional plasticity plays a central role in responding to proinflammatory stimuli. The molecular basis underlying the dynamic phenotypic activation of macrophages, however, remains incompletely understood. In this article, we report that SIRPα is a chief negative regulator of proinflammatory macrophage polarization. In response to TLR agonists, proinflammatory cytokines, or canonical M1 stimulation, Src family kinases (SFK) excluding Lyn phosphorylate SIRPα ITIMs, leading to the preferential recruitment and activation of SHP-1, but not SHP-2. Solely extracellular ligation of SIRPα by CD47 does not greatly induce phosphorylation of SIRPα ITIMs, but it enhances proinflammatory stimuli-induced SIRPα phosphorylation. Examination of downstream signaling elicited by IFN-γ and TLR3/4/9 agonists found that SIRPα-activated SHP-1 moderately represses STAT1, NF-κB, and MAPK signaling but markedly inhibits Akt2, resulting in dampened proinflammatory cytokine production and expression of Ag presentation machinery. Pharmacological inhibition of SHP-1 or deficiency of SIRPα conversely attenuates SIRPα-mediated inhibition and, as such, augments macrophage proinflammatory polarization that in turn exacerbates proinflammation in mouse models of type I diabetes and peritonitis. Our results reveal an SFK-SIRPα-SHP-1 mechanism that fine-tunes macrophage proinflammatory phenotypic activation via inhibition of PI3K-Akt2, which controls the transcription and translation of proinflammatory cytokines, Ag presentation machinery, and other cellular programs.


Asunto(s)
Inflamación/metabolismo , Macrófagos/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Presentación de Antígeno , Diferenciación Celular , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Citocinas/metabolismo , Inmunidad Celular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Inmunológicos/genética , Transducción de Señal , Células TH1/inmunología
3.
Nat Commun ; 12(1): 3229, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-34050181

RESUMEN

Radiotherapy (RT)-induced tumoricidal immunity is severely limited when tumors are well-established. Here, we report that depleting SIRPα on intratumoral macrophages augments efficacy of RT to eliminate otherwise large, treatment-resistant colorectal (MC38) and pancreatic (Pan02 and KPC) tumors, inducing complete abscopal remission and long-lasting humoral and cellular immunity that prevent recurrence. SIRPα-deficient macrophages activated by irradiated tumor-released DAMPs exhibit robust efficacy and orchestrate an anti-tumor response that controls late-stage tumors. Upon RT-mediated activation, intratumoral SIRPα-deficient macrophages acquire potent proinflammatory features and conduct immunogenic antigen presentation that confer a tumoricidal microenvironment highly infiltrated by tumor-specific cytotoxic T cells, NK cells and inflammatory neutrophils, but with limited immunosuppressive regulatory T cells, myeloid derived suppressor cells and post-radiation wound-healing. The results demonstrate that SIRPα is a master regulator underlying tumor resistance to RT and provide proof-of-principle for SIRPα-deficient macrophage-based therapies to treat a broad spectrum of cancers, including those at advanced stages with low immunogenicity and metastases.


Asunto(s)
Neoplasias/terapia , Tolerancia a Radiación/inmunología , Receptores Inmunológicos/metabolismo , Linfocitos T Citotóxicos/inmunología , Macrófagos Asociados a Tumores/inmunología , Alarminas/inmunología , Alarminas/metabolismo , Alarminas/efectos de la radiación , Animales , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoterapia/métodos , Masculino , Ratones , Ratones Noqueados , Neoplasias/inmunología , Neoplasias/patología , Prueba de Estudio Conceptual , Receptores Inmunológicos/genética , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/trasplante
4.
J Immunol ; 205(10): 2821-2833, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33028619

RESUMEN

A hallmark of secondary hemophagocytic lymphohistiocytosis (sHLH), a severe form of cytokine storm syndrome, is the emergence of overactivated macrophages that engulf healthy host blood cells (i.e., hemophagocytosis) and contribute to the dysregulated inflammation-driven pathology. In this study, we show that depleting SIRPα (SIRPα-/-) in mice during TLR9-driven inflammation exacerbates and accelerates the onset of fulminant sHLH, in which systemic hemophagocytosis, hypercytokinemia, consumptive cytopenias, hyperferritinemia, and other hemophagocytic lymphohistiocytosis hallmarks were apparent. In contrast, mice expressing SIRPα, including those deficient of the SIRPα ligand CD47 (CD47-/-), do not phenocopy SIRPα deficiency and fail to fully develop sHLH, albeit TLR9-inflamed wild-type and CD47-/- mice exhibited hemophagocytosis, anemia, and splenomegaly. Although IFN-γ is largely considered a driver of hemophagocytic lymphohistiocytosis pathology, IFN-γ neutralization did not preclude the precipitation of sHLH in TLR9-inflamed SIRPα-/- mice, whereas macrophage depletion attenuated sHLH in SIRPα-/- mice. Mechanistic studies confirmed that SIRPα not only restrains macrophages from acquiring a hemophagocytic phenotype but also tempers their proinflammatory cytokine and ferritin secretion by negatively regulating Erk1/2 and p38 activation downstream of TLR9 signaling. In addition to TLR9 agonists, TLR2, TLR3, or TLR4 agonists, as well as TNF-α, IL-6, or IL-17A, but not IFN-γ, similarly induced sHLH in SIRPα-/- mice but not SIRPα+ mice. Collectively, our study suggests that SIRPα plays a previously unappreciated role in sHLH/cytokine storm syndrome pathogenesis by preventing macrophages from becoming both hemophagocytic and hyperactivated under proinflammation.


Asunto(s)
Síndrome de Liberación de Citoquinas/inmunología , Interferón gamma/metabolismo , Linfohistiocitosis Hemofagocítica/inmunología , Macrófagos/inmunología , Receptores Inmunológicos/metabolismo , Animales , Antígeno CD47/genética , Antígeno CD47/metabolismo , Síndrome de Liberación de Citoquinas/genética , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/inmunología , Interferón gamma/antagonistas & inhibidores , Linfohistiocitosis Hemofagocítica/genética , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Noqueados , Receptores Inmunológicos/genética , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo
5.
JCI Insight ; 52019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30888336

RESUMEN

High autophagic activity in podocytes, terminally differentiated cells which serve as main components of the kidney filtration barrier, is essential for podocyte survival under various challenges. How podocytes maintain such a high level of autophagy, however, remains unclear. Here we report that signal regulatory protein α (SIRPα) plays a key role in promoting podocyte autophagy. Unlike other glomerular cells, podocytes strongly express SIRPα, which is, however, downregulated in patients with focal segmental glomerulosclerosis and mice with experimental nephropathy. Podocyte SIRPα levels are inversely correlated with the severity of podocyte injury and proteinuria but positively with autophagy. Compared to wild-type littermates, Sirpa-deficient mice display greater age-related podocyte injury and proteinuria and develop more rapid and severe renal injury in various models of experimental nephropathy. Mechanistically, podocyte SIRPα strongly reduces Akt/GSK-3ß/ß-catenin signaling, leading to an increase in autophagic activity. Our findings thus demonstrate a critical protective role of SIRPα in podocyte survival via maintaining autophagic activity.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Autofagia , Glomeruloesclerosis Focal y Segmentaria/patología , Podocitos/patología , Proteinuria/patología , Receptores Inmunológicos/metabolismo , Adolescente , Adulto , Anciano , Animales , Antígenos de Diferenciación/análisis , Biopsia , Modelos Animales de Enfermedad , Regulación hacia Abajo , Doxorrubicina/toxicidad , Femenino , Membrana Basal Glomerular/patología , Membrana Basal Glomerular/ultraestructura , Humanos , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Fosforilación , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Podocitos/ultraestructura , Proteinuria/inducido químicamente , Proteínas Proto-Oncogénicas c-akt/metabolismo , Puromicina Aminonucleósido/toxicidad , Receptores Inmunológicos/análisis , Receptores Inmunológicos/genética , Índice de Severidad de la Enfermedad , Transducción de Señal , Estreptozocina/toxicidad , Adulto Joven
6.
Eur J Immunol ; 48(6): 1046-1058, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29488625

RESUMEN

Although previous reports suggest that tumor-induced myeloid-derived suppressor cells (MDSC) inhibit T cells by L-arginine depletion through arginase-1 activity, we herein show that arginase-1 is neither inherently expressed in MDSC nor required for MDSC-mediated inhibition. Employing Percoll density gradients, large expansions of MDSC in the bone marrow of tumor-bearing mice were isolated and demonstrated potent inhibition in T-cell proliferation activated by TCR-ligation, Concanavalin A, PMA plus ionomycin, or IL-2. Despite demonstrating characteristic immunosuppressive capacity, these MDSC exhibit no arginase-1 expression and/or exert their inhibitory effects independent of arginase-1 activity. However, arginase-1 expression in MDSC can be induced by exposure to TCR-activated T cells or their culture medium, but not T cells activated by other means or growing tumor cells. Further investigation reveals multiple cytokines secreted by TCR-activated T cells as orchestrating two signaling-relay axes, IL-6-to-IL-4 and GM-CSF/IL-4-to-IL-10, leading to arginase-1 expression in MDSC. Specifically, IL-6 signaling increases IL-4R, enabling IL-4 to induce arginase-1 expression; similarly, GM-CSF in concert with IL-4 induces IL-10R, allowing IL-10-mediated induction. Surprisingly, our study indicates that induction of arginase-1 expression is not conducive to the critical MDSC-mediated inhibition toward T cells, which is rather dependent on direct cell contacts undiminished by PD-L1 blockade or SIRPα deficiency.


Asunto(s)
Arginasa/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Neoplasias Experimentales/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Animales , Arginasa/genética , Antígeno B7-H1/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular , Citocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Evasión Inmune , Tolerancia Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/agonistas , Receptores Inmunológicos/genética
7.
Sci Rep ; 8(1): 3994, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29507316

RESUMEN

Shiga toxin (Stx)-induced hemolytic uremic syndrome (HUS) is a life-threatening complication associated with Stx-producing Escherichia coli infection. One critical barrier of understanding HUS is how Stx transports from infected intestine to kidney to cause HUS. Passive dissemination seems unlikely, while circulating blood cells have been debated to serve as the toxin carrier. Employing a murine model of Stx2-induced HUS with LPS priming (LPS-Stx2), we investigate how Stx causes HUS and identify possible toxin carrier. We show that peripheral white blood cells (WBC), but not other blood cells or cell-free plasma, carry Stx2 in LPS-Stx2-treated mice. The capability of WBC binding to Stx2 is confirmed in brief ex vivo Stx2 incubation, and adoptively transferring these Stx2-bound WBC into mice induces HUS. Cell separation further identifies a subpopulation in the CD11b+ myeloid leukocytes not the CD11b- lymphocytes group act as the toxin carrier, which captures Stx2 upon exposure and delivers the toxin in vivo. Interestingly, LPS-induced inflammation significantly augments these leukocytes for binding to Stx2 and enhances HUS toxicity. Our results demonstrate that a specific fraction of circulating leukocytes carry Stx2 and cause HUS in vivo, and that LPS priming enhances the carrier capacity and aggravates organ damage.


Asunto(s)
Antígeno CD11b/inmunología , Síndrome Hemolítico-Urémico/etiología , Leucocitos/inmunología , Lipopolisacáridos/toxicidad , Toxina Shiga II/administración & dosificación , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Toxina Shiga II/toxicidad
8.
Eur J Immunol ; 48(3): 532-542, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29120053

RESUMEN

Myeloid-derived suppressor cells (MDSCs) promote tumor growth through, in part, inhibiting T-cell immunity. However, mechanisms underlying MDSC expansion and guidance of MDSCs toward the tumor microenvironment remain unclear. Employing Percoll density gradients, we separate bone marrow (BM) leukocytes from tumor-bearing mice into four density-increasing bands with myeloid leukocytes enriched in bands III and IV. Band III comprises monocytes and low-density granulocytes, both confirmed to be M-MDSCs and G-MDSCs, respectively, by displaying potent inhibition of T-cell proliferation. However, monocytes act as M-MDSCs not only under tumor conditions but also the healthy condition. In contrast, band IV contains non-inhibitory, mature granulocytes. Only band III G-MDSCs display significant expansion in mice bearing B16 melanoma, Lewis lung carcinoma, or MC38 colon carcinoma. The expanded G-MDSCs also show increased CXCR2 expression, which guides egress out of BM, and produce arginase-1 and ROS upon encountering antigen-activated T cells. Adoptive transfer assays demonstrate that both G-MDSCs and mature granulocytes infiltrate tumors, but only the former displays sustention and accumulation. Intratumoral administrations of granulocytes further demonstrate that G-MDSCs promote tumor growth, whereas mature granulocytes exert minimal effects, or execute powerful anti-tumor effects providing the presence of PMN activation mechanisms in the tumor microenvironment.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Receptores de Interleucina-8B/metabolismo , Microambiente Tumoral/inmunología , Animales , Antígenos Ly/metabolismo , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Separación Celular , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Granulocitos/inmunología , Granulocitos/patología , Activación de Linfocitos , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/patología , Células Supresoras de Origen Mieloide/patología , Linfocitos T/inmunología , Linfocitos T/patología
9.
J Immunol ; 197(8): 3293-3301, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27619992

RESUMEN

Chronic diseases are often associated with altered inflammatory response, leading to increased host vulnerability to new inflammatory challenges. Employing streptozotocin (STZ)-induced diabetes as a model, we further investigate mechanisms leading to enhanced neutrophil (polymorphonuclear leukocytes [PMN]) responses under hyperglycemia and compare them with those under chronic colitis. We show that, different from colitis under which the PMN response is significantly potentiated, the existence of a proinflammatory state associated with broad increases in macrophages in various organs plays a dominant role in promoting the PMN inflammatory response in diabetic mice. Studies of PMN infiltration during zymosan-induced peritonitis reveal that hyperglycemia enhances PMN recruitment not through inducing a high level of IL-17, which is the case in colitis, but through increasing F4/80+ macrophages in the peritoneal cavity, resulting in elevations of IL-6, IL-1ß, TNF-α, and CXCL1 production. Insulin reversal of hyperglycemia, but not the neutralization of IL-17, reduces peritoneal macrophage numbers and ameliorates PMN infiltration during peritonitis. Significantly increased macrophages are also observed in the liver, kidneys, and intestines under hyperglycemia, and they are attributable to exacerbated nephropathy and colitis when inflammatory conditions are induced by doxorubicin and dextran sulfate sodium, respectively. Furthermore, analyses of monocyte production and macrophage proliferation in tissues suggest that significant monocytosis of inflammatory F4/80+Gr-1+ monocytes from the spleen and macrophage proliferation in situ synergistically contribute to the increased macrophage population under hyperglycemia. In conclusion, our results demonstrate that STZ-induced hyperglycemic mice develop a systemic proinflammatory state mediated by broad infiltration of macrophages.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Hiperglucemia/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Animales , Diabetes Mellitus Tipo 1/inducido químicamente , Modelos Animales de Enfermedad , Hiperglucemia/inducido químicamente , Interleucina-17/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Neutrófilos/inmunología , Estreptozocina
10.
Proc Natl Acad Sci U S A ; 113(37): E5434-43, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27578867

RESUMEN

Rapid clearance of adoptively transferred Cd47-null (Cd47(-/-)) cells in congeneic WT mice suggests a critical self-recognition mechanism, in which CD47 is the ubiquitous marker of self, and its interaction with macrophage signal regulatory protein α (SIRPα) triggers inhibitory signaling through SIRPα cytoplasmic immunoreceptor tyrosine-based inhibition motifs and tyrosine phosphatase SHP-1/2. However, instead of displaying self-destruction phenotypes, Cd47(-/-) mice manifest no, or only mild, macrophage phagocytosis toward self-cells except under the nonobese diabetic background. Studying our recently established Sirpα-KO (Sirpα(-/-)) mice, as well as Cd47(-/-) mice, we reveal additional activation and inhibitory mechanisms besides the CD47-SIRPα axis dominantly controlling macrophage behavior. Sirpα(-/-) mice and Cd47(-/-) mice, although being normally healthy, develop severe anemia and splenomegaly under chronic colitis, peritonitis, cytokine treatments, and CFA-/LPS-induced inflammation, owing to splenic macrophages phagocytizing self-red blood cells. Ex vivo phagocytosis assays confirmed general inactivity of macrophages from Sirpα(-/-) or Cd47(-/-) mice toward healthy self-cells, whereas they aggressively attack toward bacteria, zymosan, apoptotic, and immune complex-bound cells; however, treating these macrophages with IL-17, LPS, IL-6, IL-1ß, and TNFα, but not IFNγ, dramatically initiates potent phagocytosis toward self-cells, for which only the Cd47-Sirpα interaction restrains. Even for macrophages from WT mice, phagocytosis toward Cd47(-/-) cells does not occur without phagocytic activation. Mechanistic studies suggest a PKC-Syk-mediated signaling pathway, to which IL-10 conversely inhibits, is required for activating macrophage self-targeting, followed by phagocytosis independent of calreticulin Moreover, we identified spleen red pulp to be one specific tissue that provides stimuli constantly activating macrophage phagocytosis albeit lacking in Cd47(-/-) or Sirpα(-/-) mice.


Asunto(s)
Antígeno CD47/genética , Inflamación/genética , Interleucina-10/genética , Receptores Inmunológicos/genética , Animales , Citocinas/biosíntesis , Citocinas/genética , Endocitosis/genética , Humanos , Inflamación/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Fagocitosis/genética , Proteína Quinasa C/genética , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...