Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
2.
FASEB J ; 34(6): 7540-7560, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32301538

RESUMEN

The vascular endothelium has been discovered in the past several years to be important in shaping the cellular immune response. During the immune response the vascular endothelium is constantly perturbed by biologically potent molecules, including the complement activation peptides, C3a and C5a. Despite the importance of C3a and C5a in inflammation and immunity, their role in modulating lymphocyte function via activation of vascular endothelial cells is unknown. Accordingly, we investigated the regulated expression of the C3a and C5a receptors (complement anaphylatoxin C3a receptor [C3aR] and complement anaphylatoxin C5a receptor 1 [C5aR1]) on human umbilical vascular endothelial cells (HUVECs) and examined how C3a or C5a activation of HUVECs affects the activation and polarization of lymphatic cells. Our findings demonstrated that C3a and C5a increase C3aR and C5aR1 expression by HUVECs as well as directing their cellular transmigration and spreading through transwell filters. Moreover, C3a- or C5a-stimulated endothelial cells: (1) caused activation of B-lymphoblasts with significant increase in Fas Ligand (CD95L) (FasL), CD69, and IL-R1 expression, and (2) skewed T-lymphoblast cells toward a Th1 subtype, (CD4+ /CCR5+ ) that correlated with significant increase of IFN-γ. Collectively, these data indicate that C3a and C5a signaling is important in the activation and polarization of lymphocytes as they traffic through the vascular endothelium during the immune response.


Asunto(s)
Anafilatoxinas/inmunología , Linfocitos B/inmunología , Complemento C3a/inmunología , Complemento C5a/inmunología , Péptidos/inmunología , Linfocitos T/inmunología , Células Cultivadas , Activación de Complemento/inmunología , Endotelio Vascular/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Complemento/inmunología , Transducción de Señal/inmunología
3.
J Immunol ; 203(10): 2701-2711, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31597707

RESUMEN

Infection with Listeria monocytogenes is acquired through ingestion of contaminated foods and may lead to systemic infection and possible death, with an overall 20% mortality rate. Our previous work using C5aR1-/- mice and C3aR-/- mice demonstrated that C5aR1 and C3aR both play powerful anti-inflammatory and prosurvival roles during systemic infection with L. monocytogenes In our current study, we have examined the role of the third anaphylatoxin receptor, C5aR2, in the host immune response to systemic L. monocytogenes infection. C5aR2-/- mice had significantly lower bacterial burdens in the spleens and livers on both day 1 and 3 postinfection compared with C5aR2+/+ mice. The decreased bacterial burdens in the C5aR2-/- mice correlated with less liver damage and with improved survival of CD4+ and CD8+ T cells in the spleen on day 3 postinfection compared with C5aR2+/+ mice. C5aR2-/- mice also produced significantly less G-CSF, IL-6, and MCP-1 in the serum, spleen, and liver on day 1 postinfection compared with C5aR2+/+ mice. C5aR2-/- and C5aR2+/+ mice produced similar amounts of IFN-γ in their spleens on day 1 postinfection. Purified naive splenocytes from C5aR2-/- mice produced significantly more IFN-γ and IL-12p70 during in vitro infection with L. monocytogenes compared with splenocytes from C5aR2+/+ mice in an NF-κB-dependent manner. Induction of IL-12 and IFN-γ early during infection with L. monocytogenes is protective to the host, and we believe this innate increased ability to produce more IL-12 and IFN-γ provided early protection to the C5aR2-/- mice.


Asunto(s)
Resistencia a la Enfermedad/inmunología , Interacciones Microbiota-Huesped/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Receptor de Anafilatoxina C5a/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular/inmunología , Complemento C5a/metabolismo , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Listeriosis/microbiología , Hígado/inmunología , Hígado/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Anafilatoxina C5a/genética , Bazo/inmunología , Bazo/microbiología , Bazo/patología
4.
J Immunol ; 198(8): 3237-3244, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28275134

RESUMEN

Listeria monocytogenes is an intracellular Gram-positive bacterium that induces expression of type I IFNs (IFN-α/IFN-ß) during infection. These cytokines are detrimental to the host during infection by priming leukocytes to undergo L. monocytogenes-mediated apoptosis. Our previous studies showed that C5aR1-/- and C3aR-/- mice are highly susceptible to L. monocytogenes infection as a result of increased IFN-ß-mediated apoptosis of major leukocyte cell populations, including CD4+ and CD8+ T cells. However, the mechanisms by which C3a and C5a modulate IFN-ß expression during L. monocytogenes infection were not examined in these initial investigations. Accordingly, we report in this article that C5a and C3a suppress IFN-ß production in response to L. monocytogenes via cyclic di-AMP (c-di-AMP), a secondary messenger molecule of L. monocytogenes, in J774A.1 macrophage-like cells and in bone marrow-derived dendritic cells (BMDCs). Moreover, C5a and C3a suppress IFN-ß production by acting through their respective receptors, because no inhibition was seen in C5aR1-/- or C3aR-/- BMDCs, respectively. C5a and C3a suppress IFN-ß production in a manner that is dependent on Bruton's tyrosine kinase, p38 MAPK, and TANK-binding kinase 1 (TBK1), as demonstrated by the individual use of Bruton's tyrosine kinase, p38 MAPK, and TBK1 inhibitors. Pretreatment of cells with C5a and C3a reduced the expression of the IFN-ß signaling molecules DDX41, STING, phosphorylated TBK1, and phosphorylated p38 MAPK in wild-type BMDCs following treatment with c-di-AMP. Collectively, these data demonstrate that C3a and C5a, via direct signaling through their specific receptors, suppress IFN-ß expression by modulation of a distinct innate cytosolic surveillance pathway involving DDX41, STING, and other downstream molecular targets of L. monocytogenes-generated c-di-AMP.


Asunto(s)
Complemento C3a/inmunología , Complemento C5a/inmunología , Inmunidad Innata/inmunología , Interferón beta/inmunología , Listeriosis/inmunología , Transducción de Señal/inmunología , Animales , Western Blotting , Complemento C3a/metabolismo , Complemento C5a/metabolismo , AMP Cíclico , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Interferón beta/biosíntesis , Listeria monocytogenes , Listeriosis/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
PLoS Pathog ; 12(6): e1005678, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27304426

RESUMEN

Spores of Bacillus anthracis, the causative agent of anthrax, are known to persist in the host lungs for prolonged periods of time, however the underlying mechanism is poorly understood. In this study, we demonstrated that BclA, a major surface protein of B. anthracis spores, mediated direct binding of complement factor H (CFH) to spores. The surface bound CFH retained its regulatory cofactor activity resulting in C3 degradation and inhibition of downstream complement activation. By comparing results from wild type C57BL/6 mice and complement deficient mice, we further showed that BclA significantly contributed to spore persistence in the mouse lungs and dampened antibody responses to spores in a complement C3-dependent manner. In addition, prior exposure to BclA deletion spores (ΔbclA) provided significant protection against lethal challenges by B. anthracis, whereas the isogenic parent spores did not, indicating that BclA may also impair protective immunity. These results describe for the first time an immune inhibition mechanism of B. anthracis mediated by BclA and CFH that promotes spore persistence in vivo. The findings also suggested an important role of complement in persistent infections and thus have broad implications.


Asunto(s)
Carbunco/inmunología , Factor H de Complemento/inmunología , Tolerancia Inmunológica/inmunología , Glicoproteínas de Membrana/inmunología , Esporas Bacterianas/inmunología , Animales , Carbunco/metabolismo , Bacillus anthracis/inmunología , Bacillus anthracis/metabolismo , Factor H de Complemento/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Esporas Bacterianas/metabolismo
7.
Brain ; 140(2): 353-369, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27956400

RESUMEN

Ischaemic stroke induces endogenous repair processes that include proliferation and differentiation of neural stem cells and extensive rewiring of the remaining neural connections, yet about 50% of stroke survivors live with severe long-term disability. There is an unmet need for drug therapies to improve recovery by promoting brain plasticity in the subacute to chronic phase after ischaemic stroke. We previously showed that complement-derived peptide C3a regulates neural progenitor cell migration and differentiation in vitro and that C3a receptor signalling stimulates neurogenesis in unchallenged adult mice. To determine the role of C3a-C3a receptor signalling in ischaemia-induced neural plasticity, we subjected C3a receptor-deficient mice, GFAP-C3a transgenic mice expressing biologically active C3a in the central nervous system, and their respective wild-type controls to photothrombotic stroke. We found that C3a overexpression increased, whereas C3a receptor deficiency decreased post-stroke expression of GAP43 (P < 0.01), a marker of axonal sprouting and plasticity, in the peri-infarct cortex. To verify the translational potential of these findings, we used a pharmacological approach. Daily intranasal treatment of wild-type mice with C3a beginning 7 days after stroke induction robustly increased synaptic density (P < 0.01) and expression of GAP43 in peri-infarct cortex (P < 0.05). Importantly, the C3a treatment led to faster and more complete recovery of forepaw motor function (P < 0.05). We conclude that C3a-C3a receptor signalling stimulates post-ischaemic neural plasticity and intranasal treatment with C3a receptor agonists is an attractive approach to improve functional recovery after ischaemic brain injury.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Complemento C3a/uso terapéutico , Plasticidad Neuronal/efectos de los fármacos , Administración Intranasal , Animales , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/etiología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/genética , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Lateralidad Funcional/efectos de los fármacos , Lateralidad Funcional/genética , Proteína GAP-43/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/genética , Desempeño Psicomotor/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/genética , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Sinapsinas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
8.
J Immunol ; 193(3): 1278-89, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24981453

RESUMEN

Listeria monocytogenes is a Gram-positive intracellular bacterium that is acquired through tainted food and may lead to systemic infection and possible death. Despite the importance of the innate immune system in fighting L. monocytogenes infection, little is known about the role of complement and its activation products, including the potent C3a anaphylatoxin. In a model of systemic L. monocytogenes infection, we show that mice lacking the receptor for C3a (C3aR(-/-)) are significantly more sensitive to infection compared with wild-type mice, as demonstrated by decreased survival, increased bacterial burden, and increased damage to their livers and spleens. The inability of the C3aR(-/-) mice to clear the bacterial infection was not caused by defective macrophages or by a reduction in cytokines/chemokines known to be critical in the host response to L. monocytogenes, including IFN-γ and TNF-α. Instead, TUNEL staining, together with Fas, active caspase-3, and Bcl-2 expression data, indicates that the increased susceptibility of C3aR(-/-) mice to L. monocytogenes infection was largely caused by increased L. monocytogenes-induced apoptosis of myeloid and lymphoid cells in the spleen that are required for ultimate clearance of L. monocytogenes, including neutrophils, macrophages, dendritic cells, and T cells. These findings reveal an unexpected function of C3a/C3aR signaling during the host immune response that suppresses Fas expression and caspase-3 activity while increasing Bcl-2 expression, thereby providing protection to both myeloid and lymphoid cells against L. monocytogenes-induced apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis/inmunología , Complemento C3a/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/patología , Receptores de Complemento/fisiología , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Caspasa 3/metabolismo , Inhibidores de Caspasas/farmacología , Complemento C3a/inmunología , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Listeria monocytogenes/patogenicidad , Listeriosis/genética , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/metabolismo , Células Mieloides/patología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Receptor fas/antagonistas & inhibidores , Receptor fas/biosíntesis
9.
J Immunol ; 193(10): 5099-107, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25297874

RESUMEN

Listeria monocytogenes is a major cause of mortality resulting from food poisoning in the United States. In mice, C5 has been genetically linked to host resistance to listeriosis. Despite this genetic association, it remains poorly understood how C5 and its activation products, C5a and C5b, confer host protection to this Gram-positive intracellular bacterium. In this article, we show in a systemic infection model that the major receptor for C5a, C5aR1, is required for a normal robust host immune response against L. monocytogenes. In comparison with wild-type mice, C5aR1(-/-) mice had reduced survival and increased bacterial burden in their livers and spleens. Infected C5aR1(-/-) mice exhibited a dramatic reduction in all major subsets of splenocytes, which was associated with elevated caspase-3 activity and increased TUNEL staining. Because type 1 IFN has been reported to impede the host response to L. monocytogenes through the promotion of splenocyte death, we examined the effect of C5aR1 on type 1 IFN expression in vivo. Indeed, serum levels of IFN-α and IFN-ß were significantly elevated in L. monocytogenes-infected C5aR1(-/-) mice. Similarly, the expression of TRAIL, a type 1 IFN target gene and a proapoptotic factor, was elevated in NK cells isolated from infected C5aR1(-/-) mice. Treatment of C5aR1(-/-) mice with a type 1 IFNR blocking Ab resulted in near-complete rescue of L. monocytogenes-induced mortality. Thus, these findings reveal a critical role for C5aR1 in host defense against L. monocytogenes through the suppression of type 1 IFN expression.


Asunto(s)
Interferón-alfa/genética , Interferón beta/genética , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Bazo/inmunología , Anafilatoxinas/inmunología , Animales , Anticuerpos/farmacología , Apoptosis , Carga Bacteriana , Caspasa 3/genética , Caspasa 3/inmunología , Complemento C5a/genética , Complemento C5a/inmunología , Complemento C5b/genética , Complemento C5b/inmunología , Expresión Génica , Interferón-alfa/inmunología , Interferón beta/inmunología , Listeriosis/tratamiento farmacológico , Listeriosis/microbiología , Listeriosis/mortalidad , Hígado/inmunología , Hígado/microbiología , Hígado/patología , Linfocitos/inmunología , Linfocitos/microbiología , Linfocitos/patología , Masculino , Ratones , Ratones Noqueados , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Receptores de Interferón/antagonistas & inhibidores , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Bazo/microbiología , Bazo/patología , Análisis de Supervivencia , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/inmunología
10.
Proc Natl Acad Sci U S A ; 110(23): 9439-44, 2013 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-23696668

RESUMEN

C3a is a key complement activation fragment, yet its neutrophil-expressed receptor (C3aR) still has no clearly defined role. In this study, we used a neutrophil-dependent mouse model of intestinal ischemia-reperfusion (IR) injury to explore the role of C3aR in acute tissue injuries. C3aR deficiency worsened intestinal injury, which corresponded with increased numbers of tissue-infiltrating neutrophils. Circulating neutrophils were significantly increased in C3aR(-/-) mice after intestinal ischemia, and C3aR(-/-) mice also mobilized more circulating neutrophils after granulocyte colony-stimulating factor infusion compared with WT mice, indicating a specific role for C3aR in constraining neutrophil mobilization in response to intestinal injury. In support of this role, C3aR(-/-) mice reconstituted with WT bone marrow reversed IR pathology back to WT levels. Complement C5a receptor (C5aR) antagonism in C3aR(-/-) mice also rectified the worsened pathology after intestinal IR injury but had no effect on circulating neutrophils, highlighting the opposing roles of C3a and C5a in disease pathogenesis. Finally, we found that using a potent C3a agonist to activate C3aR in vivo reduced neutrophil mobilization and ameliorated intestinal IR pathology in WT, but not C3aR(-/-), mice. This study identifies a role for C3aR in regulating neutrophil mobilization after acute intestinal injury and highlights C3aR agonism as a potential treatment option for acute, neutrophil-driven pathologies.


Asunto(s)
Intestinos/inmunología , Neutrófilos/inmunología , Receptores de Complemento/inmunología , Daño por Reperfusión/prevención & control , Animales , Apoptosis/inmunología , Citocinas/sangre , Hemoglobinas/metabolismo , Técnicas Histológicas , Intestinos/citología , Ratones , Ratones Noqueados , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Daño por Reperfusión/inmunología
11.
Stem Cells ; 32(2): 402-13, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24123810

RESUMEN

Human induced pluripotent stem cells (hiPSCs) have great therapeutic potential in repairing defective lung alveoli. However, genetic abnormalities caused by vector integrations and low efficiency in generating hiPSCs, as well as difficulty in obtaining transplantable hiPSC-derived cell types are still major obstacles. Here we report a novel strategy using a single nonviral site-specific targeting vector with a combination of Tet-On inducible gene expression system, Cre/lox P switching gene expression system, and alveolar epithelial type II cell (ATIIC)-specific Neomycin(R) transgene expression system. With this strategy, a single copy of all of the required transgenes can be specifically knocked into a site immediately downstream of ß-2-microglobulin (B2M) gene locus at a high frequency, without causing B2M dysfunction. Thus, the expression of reprogramming factors, Oct4, Sox2, cMyc, and Klf4, can be precisely regulated for efficient reprogramming of somatic cells into random integration-free or genetic mutation-free hiPSCs. The exogenous reprogramming factor transgenes can be subsequently removed after reprogramming by transient expression of Cre recombinase, and the resulting random integration-free and exogenous reprogramming factor-free hiPSCs can be selectively differentiated into a homogenous population of ATIICs. In addition, we show that these hiPSC-derived ATIICs exhibit ultrastructural characteristics and biological functions of normal ATIICs. When transplanted into bleomycin-challenged mice lungs, hiPSC-derived ATIICs efficiently remain and re-epithelialize injured alveoli to restore pulmonary function, preventing lung fibrosis and increasing survival without tumorigenic side effect. This strategy allows for the first time efficient generation of patient-specific ATIICs for possible future clinical applications.


Asunto(s)
Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Alveolos Pulmonares/metabolismo , Animales , Línea Celular , Reprogramación Celular/genética , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Integrasas/genética , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Pulmón , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factores de Transcripción SOXB1/biosíntesis , Factores de Transcripción SOXB1/genética
12.
J Infect Dis ; 209(8): 1269-78, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24273177

RESUMEN

BACKGROUND: The complement system protects against extracellular pathogens and links innate and adaptive immunity. In this study, we investigated the anaphylatoxin C3a receptor (C3aR) in Chlamydia psittaci lung infection and elucidated C3a-dependent adaptive immune mechanisms. METHODS: Survival, body weight, and clinical score were monitored in primary mouse infection and after serum transfer. Bacterial load, histology, cellular distribution, cytokines, antibodies, and lymphocytes were analyzed. RESULTS: C3aR(-/-) mice showed prolonged pneumonia with decreased survival, lower weight, and higher clinical score. Compared to wild-type mice bacterial clearance was impaired, and inflammatory parameters were increased. In lung-draining lymph nodes of C3aR(-/-) mice the total number of B cells, CD4(+) T cells, and Chlamydia-specific IFN-γ(+) (CD4(+) or CD8(+)) cells was reduced upon infection, and the mice were incapable of Chlamydia-specific immunoglobulin M or immunoglobulin G production. Performed before infection, transfer of hyperimmune serum prolonged survival of C3aR(-/-) mice. CONCLUSIONS: C3a and its receptor are critical for defense against C. psittaci in mouse lung infection. In this model, C3a acts via its receptor as immune modulator. Enhancement of specific B and T cell responses upon infection with an intracellular bacterium were identified as hitherto unknown features of C3a/C3aR. These new functions might be of general immunological importance.


Asunto(s)
Inmunidad Adaptativa/inmunología , Infecciones por Chlamydophila/prevención & control , Chlamydophila psittaci/patogenicidad , Pulmón/microbiología , Neumonía Bacteriana/prevención & control , Receptores de Complemento/fisiología , Linfocitos T/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Infecciones por Chlamydophila/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Masculino , Ratones , Neumonía Bacteriana/inmunología
13.
FASEB J ; 27(9): 3797-804, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23737250

RESUMEN

Complement is an essential component of inflammation that plays a role in ischemic brain injury. Recent reports demonstrate novel functions of complement in normal and diseased CNS, such as regulation of neurogenesis and synapse elimination. Here, we examined the role of complement-derived peptide C3a in unilateral hypoxia-ischemia (HI), a model of neonatal HI encephalopathy. HI injury was induced at postnatal day 9 (P9), and loss of hippocampal tissue was determined on P31. We compared WT mice with transgenic mice expressing C3a under the control of glial fibrillary acidic protein promoter, which express biologically active C3a only in CNS and without the requirement of a priori complement activation. Further, we injected C3a peptide into the lateral cerebral ventricle of mice lacking the C3a receptor (C3aR) and WT mice and assessed HI-induced memory impairment 41 d later. We found that HI-induced tissue loss in C3a overexpressing mice was reduced by 50% compared with WT mice. C3a peptide injected 1 h after HI protected WT but not C3aR-deficient mice against HI-induced memory impairment. Thus, C3a acting through its canonical receptor ameliorates behavioral deficits after HI injury, and C3aR is a novel therapeutic target for the treatment of neonatal HI encephalopathy.


Asunto(s)
Hipoxia-Isquemia Encefálica/metabolismo , Receptores de Complemento/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/metabolismo , Complemento C3a/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/patología , Inmunohistoquímica , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , ARN Mensajero/genética , Receptores de Complemento/deficiencia , Receptores de Complemento/genética
14.
Annu Rev Med ; 62: 95-105, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21226612

RESUMEN

Embryonic stem (ES) cells derived from preimplantation blastocysts and induced pluripotent stem (iPS) cells generated from somatic cell sources are pluripotent and capable of indefinite expansion in vitro. They provide a possible unlimited source of cells that could be differentiated into lung progenitor cells for potential clinical use in pulmonary regenerative medicine. Because of inherent difficulties in deriving endodermal cells from undifferentiated cell cultures, applications using lung epithelial cells derived from ES and iPS cells have lagged behind similar efforts devoted to other tissues, such as the heart and spinal cord. However, during the past several years, significant advances in culture, differentiation, and purification protocols, as well as in bioengineering methodologies, have fueled enthusiasm for the development of stem cell-based lung therapeutics. This article provides an overview of recent research achievements and discusses future technical challenges that must be met before the promise of stem cell applications for lung disease can be realized.


Asunto(s)
Células Madre Embrionarias/fisiología , Células Epiteliales/citología , Células Epiteliales/trasplante , Células Madre Pluripotentes Inducidas/fisiología , Enfermedades Pulmonares/terapia , Animales , Técnicas de Cultivo de Célula/métodos , Células Madre Embrionarias/citología , Células Epiteliales/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Pulmón/anatomía & histología , Pulmón/fisiología , Ratones , Ratas , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos
15.
FASEB J ; 26(9): 3680-90, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22651932

RESUMEN

C5a receptors are found in the central nervous system (CNS), on both neurons and glia. However, the origin of the C5a, which activates these receptors, is unclear. In the present study, we show that primary cultured mouse cortical neurons constitutively express C5, the precursor of C5a, and express the classical receptor for C5a, CD88. With cell ischemia caused by 12 h glucose deprivation, or oxygen-glucose deprivation (OGD), neurons demonstrated increased apoptosis, up-regulation of CD88, and increased levels of C5a in the media. Exogenous murine C5a (100 nM) added to the neuronal cultures resulted in apoptosis, without affecting cell necrosis. Pretreatment of the cells with the specific CD88 receptor antagonist PMX53 (100 nM) significantly blocked ischemia-induced apoptosis (∼50%), and neurons from CD88(-/-) mice were similarly protected. In a murine model of stroke, using middle cerebral artery occlusion (MCAO), we found that C5a levels in the brain increased; this also occurred in cerebral slice cultures exposed to OGD. CD88(-/-) mice subjected to MCAO had significantly reduced infarct volumes and improved neurological scores. Taken together, our results demonstrate that neurons in the CNS have the capability to generate C5a following ischemic stress, and this has the potential to activate their C5a receptors, with deleterious consequences.


Asunto(s)
Apoptosis , Isquemia Encefálica/patología , Complemento C5a/biosíntesis , Neuronas/metabolismo , Animales , Isquemia Encefálica/metabolismo , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/patología , Reacción en Cadena de la Polimerasa , Embarazo , Receptor de Anafilatoxina C5a/genética
16.
Nat Med ; 12(6): 682-7, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16715088

RESUMEN

Complement-mediated tissue injury in humans occurs upon deposition of immune complexes, such as in autoimmune diseases and acute respiratory distress syndrome. Acute lung inflammatory injury in wild-type and C3-/- mice after deposition of IgG immune complexes was of equivalent intensity and was C5a dependent, but injury was greatly attenuated in Hc-/- mice (Hc encodes C5). Injury in lungs of C3-/- mice and C5a levels in bronchoalveolar lavage (BAL) fluids from these mice were greatly reduced in the presence of antithrombin III (ATIII) or hirudin but were not reduced in similarly treated C3+/+ mice. Plasma from C3-/- mice contained threefold higher levels of thrombin activity compared to plasma from C3+/+ mice. There were higher levels of F2 mRNA (encoding prothrombin) as well as prothrombin and thrombin protein in liver of C3-/- mice compared to C3+/+ mice. A potent solid-phase C5 convertase was generated using plasma from either C3+/+ or C3-/- mice. Human C5 incubated with thrombin generated C5a that was biologically active. These data suggest that, in the genetic absence of C3, thrombin substitutes for the C3-dependent C5 convertase. This linkage between the complement and coagulation pathways may represent a new pathway of complement activation.


Asunto(s)
Activación de Complemento/fisiología , Complemento C3/inmunología , Complemento C5a/inmunología , Animales , Complejo Antígeno-Anticuerpo , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Complemento C3/genética , Complemento C5a/genética , Humanos , Inmunoglobulina G/inmunología , Hígado/citología , Hígado/metabolismo , Pulmón/inmunología , Pulmón/patología , Lesión Pulmonar , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Protrombina/genética , Protrombina/metabolismo , Trombina/metabolismo
17.
Front Immunol ; 14: 1149822, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37283747

RESUMEN

Dysregulated NLRP3 inflammasome activation drives a wide variety of diseases, while endogenous inhibition of this pathway is poorly characterised. The serum protein C4b-binding protein (C4BP) is a well-established inhibitor of complement with emerging functions as an endogenously expressed inhibitor of the NLRP3 inflammasome signalling pathway. Here, we identified that C4BP purified from human plasma is an inhibitor of crystalline- (monosodium urate, MSU) and particulate-induced (silica) NLRP3 inflammasome activation. Using a C4BP mutant panel, we identified that C4BP bound these particles via specific protein domains located on the C4BP α-chain. Plasma-purified C4BP was internalised into MSU- or silica-stimulated human primary macrophages, and inhibited MSU- or silica-induced inflammasome complex assembly and IL-1ß cytokine secretion. While internalised C4BP in MSU or silica-stimulated human macrophages was in close proximity to the inflammasome adaptor protein ASC, C4BP had no direct effect on ASC polymerisation in in vitro assays. C4BP was also protective against MSU- and silica-induced lysosomal membrane damage. We further provide evidence for an anti-inflammatory function for C4BP in vivo, as C4bp-/- mice showed an elevated pro-inflammatory state following intraperitoneal delivery of MSU. Therefore, internalised C4BP is an inhibitor of crystal- or particle-induced inflammasome responses in human primary macrophages, while murine C4BP protects against an enhanced inflammatory state in vivo. Our data suggests C4BP has important functions in retaining tissue homeostasis in both human and mice as an endogenous serum inhibitor of particulate-stimulated inflammasome activation.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Humanos , Ratones , Proteína de Unión al Complemento C4b/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Dióxido de Silicio/farmacología
18.
J Immunol ; 182(9): 5412-8, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19380788

RESUMEN

Complement effectors are known to contribute to host cell injury in several inflammatory diseases. Contrary to this paradigm, in this study utilizing surgical liver resection (partial hepatectomy) in various complement-deficient mice as a model, we have demonstrated that complement anaphylatoxins C3a and C5a are required for the survival of liver cells during regeneration. The mechanisms of these cytoprotective functions of complement were related to the regulation of IL-6 and TNF production or release after liver resection. Disturbances in the cytokine milieu, induced by a loss of complement activity, were found to alter prosurvival signaling, including the IL-6/STAT3 and PI3K/Akt/mammalian target of rapamycin pathways. In conclusion, this study documents functions of complement proteins as prosurvival factors that, through their interactions with cytokines, inhibit apoptotic signaling in proliferating cells of epithelial origin.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Hígado/citología , Hígado/inmunología , Animales , Muerte Celular/genética , Muerte Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Activación de Complemento/genética , Activación de Complemento/inmunología , Complemento C3/deficiencia , Complemento C3/genética , Complemento C3/fisiología , Complemento C3a/deficiencia , Complemento C3a/genética , Complemento C3a/fisiología , Complemento C5a/deficiencia , Complemento C5a/genética , Complemento C5a/fisiología , Proteínas del Sistema Complemento/deficiencia , Proteínas del Sistema Complemento/genética , Hepatectomía , Regeneración Hepática/genética , Regeneración Hepática/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
19.
J Immunol ; 182(10): 6533-9, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19414808

RESUMEN

Carboxypeptidase N (CPN) is a plasma zinc metalloprotease, which consists of two enzymatically active small subunits (CPN1) and two large subunits (CPN2) that protect the protein from degradation. Historically, CPN has been implicated as a major regulator of inflammation by its enzymatic cleavage of functionally important arginine and lysine amino acids from potent phlogistic molecules, such as the complement anaphylatoxins C3a and C5a. Because of no known complete CPN deficiencies, the biological impact of CPN in vivo has been difficult to evaluate. Here, we report the generation of a mouse with complete CPN deficiency by targeted disruption of the CPN1 gene. CPN1(-/-) mice were hypersensitive to lethal anaphylactic shock due to acute complement activation by cobra venom factor. This hypersensitivity was completely resolved in CPN1(-/-)/C5aR(-/-) but not in CPN1(-/-)/C3aR(-/-) mice. Moreover, CPN1(-/-) mice given C5a i.v., but not C3a, experienced 100% mortality. This C5a-induced mortality was reduced to 20% when CPN1(-/-) mice were treated with an antihistamine before C5a challenge. These studies describe for the first time a complete deficiency of CPN and demonstrate 1) that CPN plays a requisite role in regulating the lethal effects of anaphylatoxin-mediated shock, 2) that these lethal effects are mediated predominantly by C5a-induced histamine release, and 3) that C3a does not contribute significantly to shock following acute complement activation.


Asunto(s)
Complemento C5a/metabolismo , Lisina Carboxipeptidasa/genética , Choque/genética , Animales , Southern Blotting , Complemento C3a/inmunología , Complemento C3a/metabolismo , Complemento C5a/inmunología , Inactivadores del Complemento/toxicidad , Susceptibilidad a Enfermedades/inmunología , Venenos Elapídicos/toxicidad , Femenino , Histamina/inmunología , Histamina/metabolismo , Humanos , Lisina Carboxipeptidasa/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Choque/inmunología
20.
Mol Ther ; 18(3): 625-34, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20087316

RESUMEN

Respiratory diseases are a major cause of mortality and morbidity worldwide. Current treatments offer no prospect of cure or disease reversal. Transplantation of pulmonary progenitor cells derived from human embryonic stem cells (hESCs) may provide a novel approach to regenerate endogenous lung cells destroyed by injury and disease. Here, we examine the therapeutic potential of alveolar type II epithelial cells derived from hESCs (hES-ATIICs) in a mouse model of acute lung injury. When transplanted into lungs of mice subjected to bleomycin (BLM)-induced acute lung injury, hES-ATIICs behaved as normal primary ATIICs, differentiating into cells expressing phenotypic markers of alveolar type I epithelial cells. Without experiencing tumorigenic side effects, lung injury was abrogated in mice transplanted with hES-ATIICs, demonstrated by recovery of body weight and arterial blood oxygen saturation, decreased collagen deposition, and increased survival. Therefore, transplantation of hES-ATIICs shows promise as an effective therapeutic to treat acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Células Madre Embrionarias/citología , Células Epiteliales/citología , Alveolos Pulmonares/metabolismo , Animales , Diferenciación Celular , Línea Celular , Proliferación Celular , Humanos , Ratones , Oxígeno/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Trasplante de Células Madre/métodos , Transfección , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA