Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Sci Rep ; 11(1): 20545, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34654868

RESUMEN

Critical limb ischemia (CLI) is a life- and limb-threatening condition affecting 1-10% of humans worldwide with peripheral arterial disease. Cellular therapies, such as bone marrow-derived mesenchymal stem cells (MSCs) have been used for the treatment of CLI. However, little information is available regarding the angiogenic potency of MSCs and mast cells (MC) in angiogenesis. The aim of this study was to evaluate the ability of MCs and MSCs to induce angiogenesis in a rat model of ischemic hind limb injury on a background of a tissue engineered hydrogel scaffold. Thirty rats were randomly divided into six control and experimental groups as follows: (a) Control healthy (b) Ischemic positive control with right femoral artery transection, (c) ischemia with hydrogel scaffold, (d) ischemia with hydrogel plus MSC, (e) ischemia with hydrogel plus MC and (f) ischemia with hydrogel plus MSC and MCs. 106 of each cell type, isolated from bone marrow stroma, was injected into the transected artery used to induce hind limb ischemia. The other hind limb served as a non-ischemic control. After 14 days, capillary density, vascular diameter, histomorphometry and immunohistochemistry at the transected location and in gastrocnemius muscles were evaluated. Capillary density and number of blood vessels in the region of the femoral artery transection in animals receiving MSCs and MCs was increased compared to control groups (P < 0.05). Generally the effect of MCs and MSCs was similar although the combined MC/MSC therapy resulted in a reduced, rather than enhanced, effect. In the gastrocnemius muscle, immunohistochemical and histomorphometric observation showed a great ratio of capillaries to muscle fibers in all the cell-receiving groups (P < 0.05). The data indicates that the combination of hydrogel and cell therapy generates a greater angiogenic potential at the ischemic site than cell therapy or hydrogels alone.


Asunto(s)
Isquemia Crónica que Amenaza las Extremidades/terapia , Mastocitos/trasplante , Trasplante de Células Madre Mesenquimatosas , Neovascularización Fisiológica , Andamios del Tejido , Animales , Modelos Animales de Enfermedad , Masculino , Ratas Wistar
2.
PLoS One ; 16(6): e0252590, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34086763

RESUMEN

Conditions that resemble osteoarthritis (OA) were produced by injection of sodium monoiodoacetate (MIA) into the knee joints of mice. Bone marrow derived mast cells (BMMCs) injected into the OA knee joints enhanced spontaneous pain. Since no spontaneous pain was observed when BMMCs were injected into the knee joints of control mice that had not been treated with MIA, BMMCs should be activated within the OA knee joints and release some pain-inducible factors. Protease activated receptor-2 (PAR2) antagonist (FSLLRY-NH2) almost abolished the pain-enhancing effects of BMMCs injected into the OA knee joints, suggesting that tryptase, a mast cell protease that is capable of activating PAR2, should be released from the injected BMMCs and enhance pain through activation of PAR2. When PAR2 agonist (SLIGKV-NH2) instead of BMMCs was injected into the OA knee joints, it was also enhanced pain. Apyrase, an ATP degrading enzyme, injected into the OA knee joints before BMMCs suppressed the pain enhanced by BMMCs. We showed that purinoceptors (P2X4 and P2X7) were expressed in BMMCs and that extracellular ATP stimulated the release of tryptase from BMMCs. These observations suggest that ATP may stimulate degranulation of BMMCs and thereby enhanced pain. BMMCs injected into the OA knee joints stimulated expression of IL-1ß, IL-6, TNF-α, CCL2, and MMP9 genes in the infrapatellar fat pads, and PAR2 antagonist suppressed the stimulatory effects of BMMCs. Our study suggests that intermittent pain frequently observed in OA knee joints may be due, at least partly, to mast cells through activation of PAR2 and action of ATP, and that intraarticular injection of BMMCs into the OA knee joints may provide a useful experimental system for investigating molecular mechanisms by which pain is induced in OA knee joints.


Asunto(s)
Adenosina Trifosfato/metabolismo , Artritis Experimental/terapia , Dolor Crónico/patología , Articulación de la Rodilla/patología , Mastocitos/trasplante , Receptor PAR-2/metabolismo , Adenosina Trifosfato/análisis , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/patología , Células de la Médula Ósea/citología , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/toxicidad , Dolor Crónico/etiología , Modelos Animales de Enfermedad , Articulación de la Rodilla/metabolismo , Masculino , Mastocitos/citología , Mastocitos/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligopéptidos/administración & dosificación , Receptor PAR-2/agonistas , Receptor PAR-2/antagonistas & inhibidores , Receptores Purinérgicos/metabolismo , Líquido Sinovial/metabolismo
3.
Clin Exp Immunol ; 205(3): 306-315, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33999404

RESUMEN

Typical murine models of allergic inflammation are induced by the combination of ovalbumin and aluminum hydroxide. However, accumulating evidence indicates that, in models of asthma and atopic dermatitis, allergic inflammation can be generated in the absence of aluminum hydroxide. Moreover, co-administration of Staphylococcus aureus enterotoxin B with ovalbumin can enhance inflammation. The objective of this study was to establish a rapid and mast cell-dependent murine model of allergic inflammation by inducing allergic peritonitis using ovalbumin and S. aureus enterotoxin B. Allergic peritonitis was induced in C57BL/6 mice by subcutaneous sensitization and intraperitoneal challenge with ovalbumin and S. aureus enterotoxin B. Disease characteristics were assessed by flow cytometry, enzyme-linked immunosorbent assay (ELISA), trypan blue exclusion and colorimetric assays. The time-course of the allergic peritonitis revealed a peak of peritoneal inflammation 48 h after challenge, as assessed by total cells and eosinophil counts. The decrease of cell numbers started 96 h post-challenge, with complete clearance within 168 h. Moreover, significantly higher levels of tryptase and increased vascular permeability were found 30 min following challenge. Allergic inflammation induction by ovalbumin and S. aureus enterotoxin B was impaired in mast cell-deficient mice and partially restored by mice reconstitution with bone marrow-derived mast cells, indicating the mast cell role in this model. We present a novel model of allergic peritonitis that is mast cell-dependent, simple and robust. Moreover, the use of S. aureus enterotoxin B better resembles human allergic inflammation, which is known to be characterized by the colonization of S. aureus.


Asunto(s)
Asma/inmunología , Dermatitis Atópica/inmunología , Mastocitos/inmunología , Peritonitis/inmunología , Animales , Modelos Animales de Enfermedad , Enterotoxinas/inmunología , Femenino , Inmunización/métodos , Inmunoglobulina E/sangre , Inflamación/inmunología , Inflamación/patología , Masculino , Mastocitos/trasplante , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Peritonitis/patología , Staphylococcus aureus/metabolismo
4.
J Invest Surg ; 33(2): 123-133, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29869922

RESUMEN

Study Objective: We investigated the effect of bone marrow mesenchymal stem cells (BMMSCs) and chicken embryo extract (CEE), alone and in combination, on tissue viability of skin flaps, and mast cells (MCs), in an experimental random skin flap (RSF) rat model. Materials and Methods: A 30 mm × 80 mm RSF was made on the dorsum of each of the forty rats, which were then divided into four groups. One group did not receive any treatment and served as the control, the second group received BMMSCs, the third group received CEE + BMMSCs, and the fourth group received CEE. For BMMSC treatment, 6 × 109 BMMSCs were injected into twelve separate injection sites of each flap. Seven days after RSF surgery, the remaining viable part of each flap was measured and examined to determine the number of blood vessels, MCs, and degranulated MCs. Results: The CEE, CEE + BMMSC, and BMMSC groups displayed significantly higher levels of flap viability (ANOVA test: p = 0.000; LSD test for all groups: p = 0.000), and a greater number of vessels (ANOVA test: p = 0.000; LSD test: p = 0.000, 0.002, and 0.012, respectively), compared with the control group. The flap viability was poorer in the BMMSC group than in the CEE and CEE + BMMSC groups. The BMMSC group also had a greater number of degranulated and total MCs, compared with the CEE and CEE + BMMSC groups. Conclusions: We observed biostimulatory effects of BMMSCs, CEE, and CEE + BMMSCs on flap viability and vessel numbers, compared to the control group. MCs produced in response to BMMSC treatment have an inhibitory effect on the RSFs survival in an ischemic tissue model.


Asunto(s)
Mastocitos/trasplante , Trasplante de Células Madre Mesenquimatosas , Daño por Reperfusión/prevención & control , Colgajos Quirúrgicos/trasplante , Extractos de Tejidos/administración & dosificación , Animales , Células Cultivadas , Embrión de Pollo , Modelos Animales de Enfermedad , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Humanos , Masculino , Mastocitos/inmunología , Ratas , Daño por Reperfusión/inmunología , Colgajos Quirúrgicos/irrigación sanguínea
5.
Cell Death Dis ; 9(12): 1173, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518915

RESUMEN

Mast cells (MCs) are potent tissue-resident immune cells that are distributed in the intraepithelial space of the intestine and have been implicated in regulating immune homeostasis and coordinating epithelial responses in inflamed mucosa of inflammatory bowel disease (IBD). IL-33 functions as an endogenous danger signal or alarmin in inflamed intestine segments. MCs highly express the IL-33 receptor ST2. However, the mechanisms underlying the immune regulation of MC-dependent IL-33/ST2 signaling at the barrier surface of the intestine remain largely unknown. We confirmed that MCs are required for the effective resolution of tissue damage using an experimental colitis model that allows for conditional ablation of MCs. After elucidating the IL-33 signaling involved in MC activity in the context of intestinal inflammation, we found that the function of restricted IL-33/ST2 signaling by MCs was consistent with an MC deficiency in response to the breakdown of the epithelial barrier. We observed that a tissue environment with a spectrum of protective cytokines was orchestrated by MC-dependent IL-33/ST2 signaling. Given the significant downregulation of IL-22 and IL-13 due to the loss of MC-dependent IL-33/ST2 signaling and their protective functions in inflammation settings, induction of IL-22 and IL-13 may be responsible for an immune network favorable to mucosal repair. Collectively, our data showed an important feedback loop in which cytokine cues from damaged epithelia activate MCs to regulate tissue environments essential for MC-dependent restoration of epithelial barrier function and maintenance of tissue homeostasis.


Asunto(s)
Colitis/inmunología , Células Epiteliales/inmunología , Inmunidad Mucosa , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-33/inmunología , Mastocitos/inmunología , Animales , Comunicación Celular/inmunología , Colitis/inducido químicamente , Colitis/genética , Colitis/mortalidad , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Sulfato de Dextran/administración & dosificación , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Retroalimentación Fisiológica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-33/genética , Interleucinas/genética , Interleucinas/inmunología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mastocitos/efectos de los fármacos , Mastocitos/patología , Mastocitos/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Permeabilidad , Remisión Espontánea , Transducción de Señal , Análisis de Supervivencia , Interleucina-22
6.
Circ Res ; 121(8): 941-950, 2017 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-28739590

RESUMEN

RATIONALE: Deep vein thrombosis (DVT) and its complication pulmonary embolism have high morbidity reducing quality of life and leading to death. Cellular mechanisms of DVT initiation remain poorly understood. OBJECTIVE: We sought to determine the role of mast cells (MCs) in DVT initiation and validate MCs as a potential target for DVT prevention. METHODS AND RESULTS: In a mouse model, DVT was induced by partial ligation (stenosis) of the inferior vena cava. We demonstrated that 2 strains of mice deficient for MCs were completely protected from DVT. Adoptive transfer of in vitro differentiated MCs restored thrombosis. MCs were present in the venous wall, and the number of granule-containing MCs decreased with thrombosis. Pharmacological depletion of MCs granules or prevention of MC degranulation also reduced DVT. Basal plasma levels of von Willebrand factor and recruitment of platelets to the inferior vena cava wall after DVT induction were reduced in MC-deficient mice. Stenosis application increased plasma levels of soluble P-selectin in wild-type but not in MC-deficient mice. MC releasate elevated ICAM-1 (intercellular adhesion molecule-1) expression on HUVEC (human umbilical vein endothelial cells) in vitro. Topical application of compound 48/80, an MC secretagogue, or histamine, a Weibel-Palade body secretagogue from MCs, potentiated DVT in wild-type mice, and histamine restored thrombosis in MC-deficient animals. CONCLUSIONS: MCs exacerbate DVT likely through endothelial activation and Weibel-Palade body release, which is, at least in part, mediated by histamine. Because MCs do not directly contribute to normal hemostasis, they can be considered potential targets for prevention of DVT in humans.


Asunto(s)
Coagulación Sanguínea , Degranulación de la Célula , Histamina/metabolismo , Mastocitos/metabolismo , Vena Cava Inferior/metabolismo , Trombosis de la Vena/metabolismo , Traslado Adoptivo , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Degranulación de la Célula/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Fibrinolíticos/farmacología , Predisposición Genética a la Enfermedad , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Ligadura , Mastocitos/efectos de los fármacos , Mastocitos/trasplante , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Proteínas Proto-Oncogénicas c-kit/deficiencia , Proteínas Proto-Oncogénicas c-kit/genética , Selenoproteína P/metabolismo , Transducción de Señal , Vena Cava Inferior/efectos de los fármacos , Vena Cava Inferior/cirugía , Trombosis de la Vena/sangre , Trombosis de la Vena/genética , Trombosis de la Vena/prevención & control , Cuerpos de Weibel-Palade/metabolismo , Factor de von Willebrand/metabolismo
7.
Injury ; 48(7): 1480-1485, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28532897

RESUMEN

BACKGROUND: Employment of regenerative properties of cells at the service of nerve repair has been initiated during recent decades. Effects of local transplantation of bone marrow-derived mast cells on peripheral nerve regeneration were studied using a rat sciatic nerve transection model. MATERIALS AND METHODS: A 10-mm sciatic nerve defect was bridged using a conduit chitosan-based hybrid conduit filled with BMMCs in BMMC group. In positive control group (Pos), the conduit was filled with phosphate-buffered saline alone. The regenerated nerve fibers were studied within 12 weeks after surgery. In sham-operated group, the sciatic nerve was only exposed and manipulated. In negative control (Neg) a 10-mm sciatic nerve defect was created and the nerve stumps were sutured to the adjacent muscles. The regenerated nerve fibers were studied functionally, biomechanically, histologically and immunohiscochemically. RESULTS: Functional and biomechanical studies confirmed faster recovery of regenerated axons in BMMCs transplanted animals compared to Pos group (p<0.05). Morphometric indices of the regenerated fibers showed that the number and diameter of the myelinated fibers were significantly higher in BMMCs transplanted animals than in Pos group (p<0.05). In immunohistochemistry, location of reactions to S-100 in BMMCs transplanted animals was clearly more positive than that in Pos group. CONCLUSIONS: BMMCs transplantation could be considered as a readily accessible source of cells that could improve functional recovery of transected sciatic nerve.


Asunto(s)
Mastocitos/trasplante , Regeneración Nerviosa/fisiología , Nervio Ciático/lesiones , Nervio Ciático/fisiología , Animales , Materiales Biocompatibles/farmacología , Quitosano/farmacología , Masculino , Mastocitos/citología , Modelos Animales , Regeneración Nerviosa/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Nervio Ciático/cirugía
8.
Int Immunol ; 29(5): 235-242, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28431112

RESUMEN

Mast cells (MCs) play pivotal roles in allergic reactions and the host defense against microbial infection through the IgE-dependent and IgE-independent signaling pathways. MC lines that can be analyzed both in vitro and in vivo would be useful for the study of MC-dependent immune responses. Here, we investigated the functional characteristics of a mouse embryonic stem cell-derived MC-like cell line, MEDMC-BRC6. The cell line expressed FcεRI and c-Kit and showed degranulation and production of inflammatory cytokines and chemokines, including TNF-α, IL-6 and MCP-1, upon cross-linking FcεRI with IgE. These cytokines and chemokines were also produced by the cell line by stimulation of TLR2 and TLR4. MEDMC-BRC6 survived in the peritoneal cavity and the ear skin for at least 6 months after the transfer into genetically compatible MC-deficient KitW-sh/W-sh mice, in which systemic anaphylaxis was successfully induced. Thus, MEDMC-BRC6 cells represent a potent tool for investigating the functions of MCs in vitro and in vivo.


Asunto(s)
Anafilaxia/inmunología , Línea Celular/metabolismo , Mastocitos/metabolismo , Células Madre Embrionarias de Ratones/citología , Traslado Adoptivo , Animales , Degranulación de la Célula , Diferenciación Celular , Quimiocina CCL2/metabolismo , Inmunoglobulina E/inmunología , Interleucina-6/metabolismo , Mastocitos/citología , Mastocitos/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas Proto-Oncogénicas c-kit/genética , Receptores de IgG/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Hepatology ; 65(6): 1991-2004, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28120369

RESUMEN

Activated mast cells (MCs) release histamine (HA) and MCs infiltrate the liver following bile duct ligation (BDL), increasing intrahepatic bile duct mass (IBDM) and fibrosis. We evaluated the effects of BDL in MC-deficient (KitW-sh ) mice. Wild-type (WT) and KitW-sh mice were subjected to sham or BDL for up to 7 days and KitW-sh mice were injected with cultured mast cells or 1× phosphate-buffered saline (PBS) before collecting serum, liver, and cholangiocytes. Liver damage was assessed by hematoxylin and eosin and alanine aminotransferase levels. IBDM was detected by cytokeratin-19 expression and proliferation by Ki-67 immunohistochemistry (IHC). Fibrosis was detected by IHC, hydroxyproline content, and by qPCR for fibrotic markers. Hepatic stellate cell (HSC) activation and transforming growth factor-beta 1 (TGF-ß1) expression/secretion were evaluated. Histidine decarboxylase (HDC) and histamine receptor (HR) expression were detected by qPCR and HA secretion by enzymatic immunoassay. To evaluate vascular cells, von Willebrand factor (vWF) and vascular endothelial growth factor (VEGF)-C expression were measured. In vitro, cultured HSCs were stimulated with cholangiocyte supernatants and alpha-smooth muscle actin levels were measured. BDL-induced liver damage was reduced in BDL KitW-sh mice, whereas injection of MCs did not mimic BDL-induced damage. In BDL KitW-sh mice, IBDM, proliferation, HSC activation/fibrosis, and TGF-ß1 expression/secretion were decreased. The HDC/HA/HR axis was ablated in sham and BDL KitW-sh mice. vWF and VEGF-C expression decreased in BDL KitW-sh mice. In KitW-sh mice injected with MCs, IBDM, proliferation, fibrosis, and vascular cell activation increased. Stimulation with cholangiocyte supernatants from BDL WT or KitW-sh mice injected with MCs increased HSC activation, which decreased with supernatants from BDL KitW-sh mice. CONCLUSION: MCs promote hyperplasia, fibrosis, and vascular cell activation. Knockout of MCs decreases BDL-induced damage. Modulation of MCs may be important in developing therapeutics for cholangiopathies. (Hepatology 2017;65:1991-2004).


Asunto(s)
Enfermedades de las Vías Biliares/patología , Cirrosis Hepática/patología , Hígado/lesiones , Mastocitos/trasplante , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Conductos Biliares Intrahepáticos/cirugía , Enfermedades de las Vías Biliares/fisiopatología , Biopsia con Aguja , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hiperplasia/patología , Inmunohistoquímica , Ligadura/métodos , Hígado/patología , Masculino , Mastocitos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria , Valores de Referencia
10.
J Allergy Clin Immunol ; 139(2): 584-596.e10, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27555460

RESUMEN

BACKGROUND: Conflicting results have been obtained regarding the roles of Fc receptors and effector cells in models of active systemic anaphylaxis (ASA). In part, this might reflect the choice of adjuvant used during sensitization because various adjuvants might differentially influence the production of particular antibody isotypes. OBJECTIVE: We developed an "adjuvant-free" mouse model of ASA and assessed the contributions of components of the "classical" and "alternative" pathways in this model. METHODS: Mice were sensitized intraperitoneally with ovalbumin at weekly intervals for 6 weeks and challenged intraperitoneally with ovalbumin 2 weeks later. RESULTS: Wild-type animals had immediate hypothermia and late-phase intraperitoneal inflammation in this model. These features were reduced in mice lacking the IgE receptor FcεRI, the IgG receptor FcγRIII or the common γ-chain FcRγ. FcγRIV blockade resulted in a partial reduction of inflammation without any effect on hypothermia. Depletion of monocytes/macrophages with clodronate liposomes significantly reduced the hypothermia response. By contrast, depletion of neutrophils or basophils had no significant effects in this ASA model. Both the hypothermia and inflammation were dependent on platelet-activating factor and histamine and were reduced in 2 types of mast cell (MC)-deficient mice. Finally, engraftment of MC-deficient mice with bone marrow-derived cultured MCs significantly exacerbated the hypothermia response and restored inflammation to levels similar to those observed in wild-type mice. CONCLUSION: Components of the classical and alternative pathways contribute to anaphylaxis in this adjuvant-free model, with key roles for MCs and monocytes/macrophages.


Asunto(s)
Anafilaxia/inmunología , Movimiento Celular , Hipotermia/inmunología , Leucocitos/inmunología , Macrófagos/inmunología , Mastocitos/inmunología , Adyuvantes Inmunológicos , Animales , Células Cultivadas , Vía Alternativa del Complemento , Vía Clásica del Complemento , Modelos Animales de Enfermedad , Humanos , Inmunización , Mastocitos/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de IgE/genética , Receptores de IgE/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo
11.
J Innate Immun ; 9(1): 94-108, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27806369

RESUMEN

Mucosal surfaces are protected from infection by both structural and sentinel cells, such as mast cells. The mast cell's role in antiviral responses is poorly understood; however, they selectively recruit natural killer (NK) cells following infection. Here, the ability of virus-infected mast cells to enhance NK cell functions was examined. Cord blood-derived human mast cells infected with reovirus (Reo-CBMC) and subsequent mast cell products were used for the stimulation of human NK cells. NK cells upregulated the CD69 molecule and cytotoxicity-related genes, and demonstrated increased cytotoxic activity in response to Reo-CBMC soluble products. NK cell interferon (IFN)-γ production was also promoted in the presence of interleukin (IL)-18. In vivo, SCID mice injected with Reo-CBMC in a subcutaneous Matrigel model, could recruit and activate murine NK cells, a property not shared by normal human fibroblasts. Soluble products of Reo-CBMC included IL-10, TNF, type I and type III IFNs. Blockade of the type I IFN receptor abrogated NK cell activation. Furthermore, reovirus-infected mast cells expressed multiple IFN-α subtypes not observed in reovirus-infected fibroblasts or epithelial cells. Our data define an important mast cell IFN response, not shared by structural cells, and a subsequent novel mast cell-NK cell immune axis in human antiviral host defense.


Asunto(s)
Inmunidad Mucosa , Células Asesinas Naturales/inmunología , Mastocitos/inmunología , Orthoreovirus de los Mamíferos/inmunología , Infecciones por Reoviridae/inmunología , Animales , Células Cultivadas , Citotoxicidad Inmunológica , Sangre Fetal/citología , Humanos , Interferones/metabolismo , Interleucina-18/metabolismo , Mastocitos/trasplante , Mastocitos/virología , Ratones , Ratones SCID , Especificidad de Órganos , Comunicación Paracrina , Receptor de Interferón alfa y beta/antagonistas & inhibidores
12.
Oncotarget ; 7(50): 82985-83000, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27783996

RESUMEN

Systemic mastocytosis are rare neoplasms characterized by accumulation of mast cells in at least one internal organ. The majority of systemic mastocytosis patients carry KIT D816V mutation, which activates constitutively the KIT receptor. Patient with advanced forms of systemic mastocytosis, such as aggressive systemic mastocytosis or mast cell leukemia, are poorly treated to date. Unfortunately, the lack of in vivo models reflecting KIT D816V+ advanced disease hampers pathophysiological studies and preclinical development of new therapies for such patients. Here, we describe a new in vivo model of KIT D816V+ advanced systemic mastocytosis developed by transplantation of the human ROSAKIT D816V-Gluc mast cell line in NOD-SCID IL-2R γ-/- mice, using Gaussia princeps luciferase as a reporter. Intravenous injection of ROSAKIT D816V-Gluc cells led, in 4 weeks, to engraftment in all injected primary recipient mice. Engrafted cells were found at high levels in bone marrow, and at lower levels in spleen, liver and peripheral blood. Disease progression was easily monitored by repeated quantification of Gaussia princeps luciferase activity in peripheral blood. This quantification evidenced a linear relationship between the number of cells injected and the neoplastic mast cell burden in mice. Interestingly, the secondary transplantation of ROSAKIT D816V-Gluc cells increased their engraftment capability. To conclude, this new in vivo model mimics at the best the features of human KIT D816V+ advanced systemic mastocytosis. In addition, it is a unique and convenient tool to study the kinetics of the disease and the potential in vivo activity of new drugs targeting neoplastic mast cells.


Asunto(s)
Genes Reporteros , Luciferasas/genética , Mastocitos/trasplante , Mastocitosis Sistémica/genética , Mutación , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Luciferasas/biosíntesis , Luciferasas/sangre , Mastocitos/efectos de los fármacos , Mastocitos/enzimología , Mastocitos/patología , Mastocitosis Sistémica/tratamiento farmacológico , Mastocitosis Sistémica/enzimología , Mastocitosis Sistémica/patología , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factores de Tiempo , Transfección
13.
Nat Commun ; 7: 13198, 2016 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-27786273

RESUMEN

Cross-linkage of the high-affinity immunoglobulin E (IgE) receptor (FcɛRI) on mast cells by antigen ligation has a critical role in the pathology of IgE-dependent allergic disorders, such as anaphylaxis and asthma. Restraint of intracellular signal transduction pathways that promote release of mast cell-derived pro-inflammatory mediators is necessary to dampen activation and restore homoeostasis. Here we show that the ligase Nedd4-2 and the adaptor Ndfip1 (Nedd4 family interacting protein 1) limit the intensity and duration of IgE-FcɛRI-induced positive signal transduction by ubiquitinating phosphorylated Syk, a tyrosine kinase that is indispensable for downstream FcɛRI signalosome activity. Importantly, loss of Nedd4-2 or Ndfip1 in mast cells results in exacerbated and prolonged IgE-mediated cutaneous anaphylaxis in vivo. Our findings reveal an important negative regulatory function for Nedd4-2 and Ndfip1 in IgE-dependent mast cell activity.


Asunto(s)
Proteínas Portadoras/inmunología , Inmunoglobulina E/inmunología , Mastocitos/inmunología , Proteínas de la Membrana/inmunología , Ubiquitina-Proteína Ligasas Nedd4/inmunología , Traslado Adoptivo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Inmunoglobulina E/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Masculino , Mastocitos/metabolismo , Mastocitos/trasplante , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ubiquitina-Proteína Ligasas Nedd4/genética , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Anafilaxis Cutánea Pasiva/genética , Anafilaxis Cutánea Pasiva/inmunología , Receptores de IgE/inmunología , Receptores de IgE/metabolismo , Quinasa Syk/inmunología , Quinasa Syk/metabolismo
14.
J Cell Mol Med ; 19(12): 2751-62, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26471858

RESUMEN

Multiple mechanisms contribute to progressive cardiac dysfunction after myocardial infarction (MI) and inflammation is an important mediator. Mast cells (MCs) trigger inflammation after MI by releasing bio-active factors that contribute to healing. c-Kit-deficient (Kit(W/W-v) ) mice have dysfunctional MCs and develop severe ventricular dilatation post-MI. We explored the role of MCs in post-MI repair. Mouse wild-type (WT) and Kit(W/W-v) MCs were obtained from bone marrow (BM). MC effects on fibroblasts were examined in vitro by proliferation and gel contraction assays. MCs were implanted into infarcted mouse hearts and their effects were evaluated using molecular, cellular and cardiac functional analyses. In contrast to WT, Kit(W/W-v) MC transplantation into Kit(W/W-v) mice did not improve cardiac function or scar size post-MI. Kit(W/W-v) MCs induced significantly reduced fibroblast proliferation and contraction compared to WT MCs. MC influence on fibroblast proliferation was Basic fibroblast growth factor (bFGF)-dependent and MC-induced fibroblast contractility functioned through transforming growth factor (TGF)-ß. WT MCs transiently rescue cardiac function early post-MI, but the benefits of BM cell implantation lasted longer. MCs induced increased inflammation compared to the BM-injected mice, with increased neutrophil infiltration and infarct tumour necrosis factor-α (TNF-α) concentration. This augmented inflammation was followed by increased angiogenesis and myofibroblast formation and reduced scar size at early time-points. Similar to the functional data, these beneficial effects were transient, largely vanishing by day 28. Dysfunctional Kit(W/W-v) MCs were unable to rescue cardiac function post-MI. WT MC implantation transiently enhanced angiogenesis and cardiac function. These data suggest that increased inflammation is beneficial to cardiac repair, but these effects are not persistent.


Asunto(s)
Inflamación/metabolismo , Mastocitos/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/metabolismo , Citometría de Flujo , Inflamación/fisiopatología , Inflamación/terapia , Mastocitos/trasplante , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Miocardio/patología , Miofibroblastos/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
15.
Immunity ; 43(4): 788-802, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26410628

RESUMEN

Experimental IgE-mediated food allergy depends on intestinal anaphylaxis driven by interleukin-9 (IL-9). However, the primary cellular source of IL-9 and the mechanisms underlying the susceptibility to food-induced intestinal anaphylaxis remain unclear. Herein, we have reported the identification of multifunctional IL-9-producing mucosal mast cells (MMC9s) that can secrete prodigious amounts of IL-9 and IL-13 in response to IL-33, and mast cell protease-1 (MCPt-1) in response to antigen and IgE complex crosslinking, respectively. Repeated intragastric antigen challenge induced MMC9 development that required T cells, IL-4, and STAT6 transcription factor, but not IL-9 signals. Mice ablated of MMC9 induction failed to develop intestinal mastocytosis, which resulted in decreased food allergy symptoms that could be restored by adoptively transferred MMC9s. Finally, atopic patients that developed food allergy displayed increased intestinal expression of Il9- and MC-specific transcripts. Thus, the induction of MMC9s is a pivotal step to acquire the susceptibility to IgE-mediated food allergy.


Asunto(s)
Hipersensibilidad a los Alimentos/inmunología , Inmunoglobulina E/inmunología , Interleucina-9/metabolismo , Mucosa Intestinal/inmunología , Mastocitos/inmunología , Mastocitosis/inmunología , Traslado Adoptivo , Anafilaxia/etiología , Anafilaxia/inmunología , Animales , Secuencia de Bases , Células de la Médula Ósea/citología , Linaje de la Célula , Quimasas/biosíntesis , Quimasas/genética , Diarrea/etiología , Diarrea/inmunología , Susceptibilidad a Enfermedades , Duodeno/inmunología , Duodeno/patología , Hipersensibilidad a los Alimentos/etiología , Hipersensibilidad a los Alimentos/patología , Humanos , Hipersensibilidad Inmediata/complicaciones , Interleucina-9/biosíntesis , Interleucina-9/genética , Interleucinas/biosíntesis , Interleucinas/metabolismo , Interleucinas/fisiología , Mastocitos/metabolismo , Mastocitos/trasplante , Mastocitosis/patología , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Ovalbúmina/toxicidad , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factor de Transcripción STAT6/fisiología , Especificidad de la Especie , Linfocitos T/inmunología
16.
Cancer Res ; 75(18): 3760-70, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26206557

RESUMEN

Mast cells (MC) are immune cells located next to the intestinal epithelium with regulatory function in maintaining the homeostasis of the mucosal barrier. We have investigated MC activities in colon inflammation and cancer in mice either wild-type (WT) or MC-deficient (Kit(W-sh)) reconstituted or not with bone marrow-derived MCs. Colitis was chemically induced with dextran sodium sulfate (DSS). Tumors were induced by administering azoxymethane (AOM) intraperitoneally before DSS. Following DSS withdrawal, Kit(W-sh) mice showed reduced weight gain and impaired tissue repair compared with their WT littermates or Kit(W-sh) mice reconstituted with bone marrow-derived MCs. MCs were localized in areas of mucosal healing rather than damaged areas where they degraded IL33, an alarmin released by epithelial cells during tissue damage. Kit(W-sh) mice reconstituted with MC deficient for mouse mast cell protease 4 did not restore normal mucosal healing or reduce efficiently inflammation after DSS withdrawal. In contrast with MCs recruited during inflammation-associated wound healing, MCs adjacent to transformed epithelial cells acquired a protumorigenic profile. In AOM- and DSS-treated WT mice, high MC density correlated with high-grade carcinomas. In similarly treated Kit(W-sh) mice, tumors were less extended and displayed lower histologic grade. Our results indicate that the interaction of MCs with epithelial cells is dependent on the inflammatory stage, and on the activation of the tissue repair program. Selective targeting of MCs for prevention or treatment of inflammation-associated colon cancer should be timely pondered to allow tissue repair at premalignant stages or to reduce aggressiveness at the tumor stage.


Asunto(s)
Carcinoma/inmunología , Colitis/inmunología , Neoplasias del Colon/inmunología , Mucosa Intestinal/fisiología , Mastocitos/fisiología , Regeneración/inmunología , Animales , Animales Congénicos , Azoximetano/toxicidad , Carcinoma/inducido químicamente , Carcinoma/patología , Recuento de Células , Transformación Celular Neoplásica/inmunología , Células Cultivadas , Colitis/inducido químicamente , Colitis/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/patología , Sulfato de Dextran/toxicidad , Células Epiteliales/patología , Humanos , Enfermedades Inflamatorias del Intestino/patología , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33/fisiología , Mastocitos/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Proteínas Proto-Oncogénicas c-kit/deficiencia , Proteínas Proto-Oncogénicas c-kit/genética , Receptores de Interleucina/fisiología , Serina Endopeptidasas/deficiencia , Especificidad de la Especie , Organismos Libres de Patógenos Específicos
17.
J Vis Exp ; (99): e52753, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26068439

RESUMEN

Mast cells (MCs) are hematopoietic cells which reside in various tissues, and are especially abundant at sites exposed to the external environment, such as skin, airways and gastrointestinal tract. Best known for their detrimental role in IgE-dependent allergic reactions, MCs have also emerged as important players in host defense against venom and invading bacteria and parasites. MC phenotype and function can be influenced by microenvironmental factors that may differ according to anatomic location and/or based on the type or stage of development of immune responses. For this reason, we and others have favored in vivo approaches over in vitro methods to gain insight into MC functions. Here, we describe methods for the generation of mouse bone marrow-derived cultured MCs (BMCMCs), their adoptive transfer into genetically MC-deficient mice, and the analysis of the numbers and distribution of adoptively transferred MCs at different anatomical sites. This method, named the 'mast cell knock-in' approach, has been extensively used over the past 30 years to assess the functions of MCs and MC-derived products in vivo. We discuss the advantages and limitations of this method, in light of alternative approaches that have been developed in recent years.


Asunto(s)
Traslado Adoptivo/métodos , Mastocitos/citología , Animales , Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula/métodos , Trasplante de Células/métodos , Femenino , Masculino , Mastocitos/trasplante , Ratones , Ratones Transgénicos , Piel/citología , Piel/inmunología
19.
Cancer Lett ; 353(1): 115-23, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25088577

RESUMEN

Gain-of-function mutations of receptor tyrosine kinase KIT play a critical role in the pathogenesis of systemic mastocytosis (SM) and gastrointestinal stromal tumors. D816V KIT mutation, found in ∼80% of SM, is resistant to the currently available tyrosine kinase inhibitors (TKIs) (e.g. imatinib mesylate). Therefore, development of promising TKIs for the treatment of D816V KIT mutation is still urgently needed. We synthesized thiazole amine compounds and chose one representative designated 126332 to investigate its effect on human mast cells expressing KIT mutations. We found 126332 inhibited the phosphorylation of KIT and its downstream signaling molecules Stat3 and Stat5. 126332 inhibited the proliferation of D816V KIT expressing cells. 126332 induced apoptosis and downregulated levels of Mcl-1 and survivin. Furthermore, 126332 inhibited the tyrosine phosphorylation of ß-catenin, inhibited ß-catenin-mediated transcription and DNA binding of TCF. Moreover, 126332 also exhibited in vivo antineoplastic activity against cells harboring D816V mutation. Our findings suggest thiazole amine compounds may be promising agents for the treatment of diseases caused by KIT mutation.


Asunto(s)
Aminas/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Mastocitos/efectos de los fármacos , Mastocitoma/tratamiento farmacológico , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/genética , Tiazoles/farmacología , Aminas/síntesis química , Animales , Antineoplásicos/síntesis química , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Genotipo , Humanos , Masculino , Mastocitos/enzimología , Mastocitos/patología , Mastocitos/trasplante , Mastocitoma/enzimología , Mastocitoma/genética , Mastocitoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Terapia Molecular Dirigida , Fenotipo , Fosforilación , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Proto-Oncogénicas c-kit/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Tiazoles/síntesis química , Factores de Tiempo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Immunol ; 193(4): 1886-94, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25015817

RESUMEN

ß-Hexosaminidase, which is generally present in the lysosome, is essential for glycoprotein metabolism in the maintenance of cell homeostasis. In mast cells (MCs), large amounts of ß-hexosaminidase are present in the granules as opposed to the lysosome, and the biological role of MC ß-hexosaminidase has yet to be fully elucidated. Therefore, we investigated the biological role of ß-hexosaminidase in MC granules. Bone marrow-derived MCs from C57BL/6 (BL/6-BMMC) or ß-hexosaminidase gene-deficient (hexb(-/-)-BMMC) mice were transplanted into MC-deficient (WBB6F1/J-Kit(W)/Kit(W-v) [W/W(v)]) mice to generate MC-reconstituted models. In asthma model experiments, no differences were observed in the symptoms of BL/6, W/W(v), BL/6-BMMC-reconstituted W/W(v), or hexb(-/-)-BMMC-reconstituted W/W(v) mice. In Staphylococcus epidermidis experimental infection model experiments, the severity of symptoms and frequency of death were markedly higher in W/W(v) and hexb(-/-)-BMMC-reconstituted W/W(v) mice than in BL/6 and BL/6-BMMC-reconstituted W/W(v) mice. The growth of S. epidermidis in an in vitro study was clearly inhibited by addition of BL/6-BMMC lysate, but not by addition of hexb(-/-)-BMMC lysate. Moreover, suppression of bacterial proliferation was completely recovered when bacteria were incubated with hexb(-/-)-BMMC lysate plus ß-hexosaminidase. Transmission electron microscopy indicated that the cell wall of S. epidermidis was heavily degraded following coincubation of bacteria with BL/6-BMMC lysate, but not following coincubation with hexb(-/-)-BMMC lysate. These findings strongly suggest that MC granule ß-hexosaminidase is crucial for defense against bacterial invasion, but is not involved in the allergic response. Our results also suggest that the bactericidal mechanism of ß-hexosaminidase involves degradation of bacterial cell wall peptidoglycan.


Asunto(s)
Gránulos Citoplasmáticos/enzimología , Mastocitos/enzimología , Mastocitos/inmunología , Infecciones Estafilocócicas/inmunología , beta-N-Acetilhexosaminidasas/metabolismo , Animales , Asma/inmunología , Degranulación de la Célula , Pared Celular/inmunología , Modelos Animales de Enfermedad , Glicoproteínas/metabolismo , Lisosomas/enzimología , Mastocitos/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peptidoglicano/inmunología , Peptidoglicano/metabolismo , Infecciones Estafilocócicas/mortalidad , Staphylococcus epidermidis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...