Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 24(2): 239-254, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36604547

RESUMEN

Metastasis is the leading cause of cancer-related deaths and myeloid cells are critical in the metastatic microenvironment. Here, we explore the implications of reprogramming pre-metastatic niche myeloid cells by inducing trained immunity with whole beta-glucan particle (WGP). WGP-trained macrophages had increased responsiveness not only to lipopolysaccharide but also to tumor-derived factors. WGP in vivo treatment led to a trained immunity phenotype in lung interstitial macrophages, resulting in inhibition of tumor metastasis and survival prolongation in multiple mouse models of metastasis. WGP-induced trained immunity is mediated by the metabolite sphingosine-1-phosphate. Adoptive transfer of WGP-trained bone marrow-derived macrophages reduced tumor lung metastasis. Blockade of sphingosine-1-phosphate synthesis and mitochondrial fission abrogated WGP-induced trained immunity and its inhibition of lung metastases. WGP also induced trained immunity in human monocytes, resulting in antitumor activity. Our study identifies the metabolic sphingolipid-mitochondrial fission pathway for WGP-induced trained immunity and control over metastasis.


Asunto(s)
Neoplasias Pulmonares , beta-Glucanos , Animales , Ratones , Humanos , Inmunidad Entrenada , Macrófagos , Lisofosfolípidos/metabolismo , Monocitos , Neoplasias Pulmonares/patología , beta-Glucanos/metabolismo , beta-Glucanos/farmacología , Microambiente Tumoral
2.
J Immunol ; 207(7): 1785-1797, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34470858

RESUMEN

Our previous work has revealed the ability of CD11b to regulate BCR signaling and control autoimmune disease in mice. However, how CD11b regulates the immune response under normal conditions remains unknown. Through the use of a CD11b knockout model on a nonautoimmune background, we demonstrated that CD11b-deficient mice have an elevated Ag-specific humoral response on immunization. Deletion of CD11b resulted in elevated low-affinity and high-affinity IgG Ab and increases in Ag-specific germinal center B cells and plasma cells (PCs). Examination of BCR signaling in CD11b-deficient mice revealed defects in association of negative regulators pLyn and CD22 with the BCR, but increases in colocalizations between positive regulator pSyk and BCR after stimulation. Using a CD11b-reporter mouse model, we identified multiple novel CD11b-expressing B cell subsets that are dynamically altered during immunization. Subsequent experiments using a cell-specific CD11b deletion model revealed this effect to be B cell intrinsic and not altered by myeloid cell CD11b expression. Importantly, CD11b expression on PCs also impacts on BCR repertoire selection and diversity in autoimmunity. These studies describe a novel role for CD11b in regulation of the healthy humoral response and autoimmunity, and reveal previously unknown populations of CD11b-expressing B cell subsets, suggesting a complex function for CD11b in B cells during development and activation.


Asunto(s)
Linfocitos B/inmunología , Antígeno CD11b/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Autoinmunidad , Antígeno CD11b/genética , Células Cultivadas , Humanos , Inmunidad Humoral , Inmunización , Inmunomodulación , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Receptores de Antígenos de Linfocitos B/genética , Transducción de Señal , Quinasa Syk/metabolismo
3.
Int J Mol Sci ; 24(17)2023 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-37686472

RESUMEN

The signal transducer and activator of transcription 3 (STAT3), which regulates multiple oncogenic processes, has been found to be constitutively activated in lymphoma, suggesting its potential as a therapeutic target. Here, we constructed an anti-CD19-N-(4-carboxycyclohexylmethyl) maleimide N-hydroxysuccinimide ester (SMCC)-protamine (CSP)-STAT3 small interfering RNA (siRNA) conjugate and demonstrated that the CSP-STAT3 siRNA conjugate could specifically bind to normal B cells and A20 lymphoma cells in vitro. It decreased the STAT3 expression in B cell lymphoma cell lines (A20, SU-DHL-2 and OCI-Ly3), resulting in reduced proliferation of lymphoma cells featured with lower S-phase and higher apoptosis. Using an A20 transplantable lymphoma model, we found that the CSP-STAT3 siRNA conjugate significantly inhibited tumor growth and weight. Ki-67, p-STAT3, STAT3, and serum IL-6 levels were all significantly reduced in A20-bearing mice treated with CSP-STAT3 siRNA. These findings indicate that specifically targeting STAT3 siRNA to B cell lymphoma cell lines can significantly decrease STAT3 activity and inhibit tumor progression in vitro and in vivo, suggesting its potential utilization for cancer treatment.


Asunto(s)
Linfoma de Células B , Factor de Transcripción STAT3 , Animales , Ratones , Proteínas Adaptadoras Transductoras de Señales , Anticuerpos , Linfocitos B , Linfoma de Células B/genética , Linfoma de Células B/terapia , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/genética
4.
Biophys J ; 121(6): 966-976, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35150619

RESUMEN

Innate immune cells detect pathogens through simultaneous stimulation of multiple receptors, but how cells use the receptor crosstalk to elicit context-appropriate responses is unclear. Here, we reveal that the inflammatory response of macrophages from FcγR-TLR2/1 crosstalk inversely depends on the ligand mobility within a model pathogen membrane. The mechanism is that FcγR and TLR2/1 form separate nanoclusters that interact at their interfaces during crosstalk. Less mobile ligands induce stronger interactions and more overlap between the receptor nanoclusters, leading to enhanced signaling. Different from the prevailing view that immune receptors colocalize to synergize their signaling, our results show that FcγR-TLR2/1 crosstalk occurs through interface interactions between non-colocalizing receptor nanoclusters, which are modulated by ligand mobility. This suggests a mechanism by which innate immune cells could use physical properties of ligands to fine-tune host responses.


Asunto(s)
Receptores de IgG , Receptor Toll-Like 2 , Ligandos , Macrófagos , Transducción de Señal
5.
J Immunol ; 204(1): 212-223, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31776206

RESUMEN

Regulation of myeloid-derived suppressor cells (MDSC) by ongoing inflammation following repeated chemotherapy remain elusive. In this study, we show that a multidose clinical regimen of gemcitabine (GEM) treatment enhances the immunosuppressive function of monocytic MDSC (M-MDSC), although tumor development is delayed in E0771 tumor-bearing mice. Accordingly, effector IFN-γ-producing CD4 and CD8 T cells are significantly decreased in the tumor microenvironment (TME) of GEM-treated mice. The conditioned medium of GEM-treated tumor cells enhances differentiation of mouse bone marrow cells and human PBMC into immunosuppressive M-MDSC. Cytokine profiling of GEM-treated tumor cells identifies GM-CSF as one of the most differentially expressed cytokines. Blockade or knockdown of GM-CSF can partially reduce immunosuppression of Ly6Chigh cells induced by GEM-conditioned medium. Knockdown of GM-CSF in tumor cells also delays tumor progression with decreased accumulation of M-MDSC in the TME. Mechanistically, enhanced production of reactive oxygen species and activation of NF-κB are observed in GEM-treated tumor cells. Treatment with the mitochondrial-targeted antioxidant and inhibitor of NF-κB signaling can abrogate GEM-induced hyperexpression of GM-CSF in E0771 cells. In addition, the phagocytic clearance of apoptotic tumor cells (efferocytosis) enhances the immunosuppressive function of bone marrow Ly6Chigh myeloid cells. Further, GEM treatment results in metabolic changes in residual tumor cells, leading to the resistance to T cell-mediated killing. Together, our results define an undesired effect of repeated GEM treatment promoting immunosuppression in TME via upregulation of GM-CSF and efferocytosis as well as deregulation of lipid metabolism in residual tumor cells.


Asunto(s)
Antígenos Ly/inmunología , Antimetabolitos Antineoplásicos/farmacología , Desoxicitidina/análogos & derivados , Inmunosupresores/farmacología , Células Mieloides/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Proliferación Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Inmunosupresores/administración & dosificación , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/inmunología , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Células Tumorales Cultivadas , Gemcitabina
6.
J Immunol ; 202(5): 1623-1634, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30665937

RESUMEN

The role of the mTOR signaling pathway in different myeloid cell subsets is poorly understood in the context of tumor development. In this study, myeloid cell-specific Raptor knockout (KO) mice were used to determine the roles of mechanistic target of rapamycin complex 1 (mTORC1) in regulating macrophage function from Lewis lung carcinoma (LLC) s.c. tumors and lung tumor metastasis. We found no difference in tumor growth between conditional Raptor KO and control mice in the s.c. tumor models, although depletion of mTORC1 decreased the immunosuppressive function of tumor-associated macrophages (TAM). Despite the decreased immunosuppressive activity of TAM, M1-like TAM differentiation was impaired in the s.c. tumor microenvironment of mTORC1 conditional Raptor KO mice due to downregulated CD115 expression on macrophages. In addition, TNF-α production by mTORC1-deficient myeloid cells was also decreased in the s.c. LLC tumors. On the contrary, disruption of mTORC1 in myeloid cells promoted lung cancer metastasis. Accordingly, immunosuppressive interstitial macrophages/metastasis-associated macrophages (CD11b+F4/80high) were accumulated in the lungs of Raptor KO mice in the LLC lung metastasis model, leading to decreased Th1 responses. Taken together, our results demonstrate that differential tumor microenvironment dictates the immunological outcomes of myeloid cells, with mTORC1 disruption leading to different tumor growth phenotypes.


Asunto(s)
Neoplasias Pulmonares/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Células Mieloides/inmunología , Microambiente Tumoral/inmunología , Animales , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Ratones Transgénicos
7.
Immunity ; 35(4): 596-610, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21982596

RESUMEN

Interleukin-23 (IL-23) and CD4(+) T helper 17 (Th17) cells are thought to be critical in psoriasis pathogenesis. Here, we report that IL-23 predominantly stimulated dermal γδ T cells to produce IL-17 that led to disease progression. Dermal γδ T cells constitutively expressed the IL-23 receptor (IL-23R) and transcriptional factor RORγt. IL-17 production from dermal γδ T cells was independent of αß T cells. The epidermal hyperplasia and inflammation induced by IL-23 were significantly decreased in T cell receptor δ-deficient (Tcrd(-/-)) and IL-17 receptor-deficient (Il17ra(-/-)) mice but occurred normally in Tcra(-/-) mice. Imiquimod-induced skin pathology was also significantly decreased in Tcrd(-/-) mice. Perhaps further promoting disease progression, IL-23 stimulated dermal γδ T cell expansion. In psoriasis patients, γδ T cells were greatly increased in affected skin and produced large amounts of IL-17. Thus, IL-23-responsive dermal γδ T cells are the major IL-17 producers in the skin and may represent a novel target for the treatment of psoriasis.


Asunto(s)
Dermatitis/inmunología , Interleucina-17/biosíntesis , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Animales , Dermatitis/patología , Humanos , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-23/biosíntesis , Interleucina-23/inmunología , Macrófagos/inmunología , Ratones , Ratones Noqueados , Fenotipo , Psoriasis/inmunología , Psoriasis/patología , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Receptores de Interleucina-17/inmunología , Linfocitos T/metabolismo
8.
J Immunol ; 196(5): 2167-80, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26810222

RESUMEN

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature myeloid cells that promote tumor progression. In this study, we demonstrated that activation of a C-type lectin receptor, dectin-1, in MDSC differentially modulates the function of different MDSC subsets. Yeast-derived whole ß-glucan particles (WGP; a ligand to engage and activate dectin-1, oral treatment in vivo) significantly decreased tumor weight and splenomegaly in tumor-bearing mice with reduced accumulation of polymorphonuclear MDSC but not monocytic MDSC (M-MDSC), and decreased polymorphonuclear MDSC suppression in vitro through the induction of respiratory burst and apoptosis. On a different axis, WGP-treated M-MDSC differentiated into F4/80(+)CD11c(+) cells in vitro that served as potent APC to induce Ag-specific CD4(+) and CD8(+) T cell responses in a dectin-1-dependent manner. Additionally, Erk1/2 phosphorylation was required for the acquisition of APC properties in M-MDSC. Moreover, WGP-treated M-MDSC differentiated into CD11c(+) cells in vivo with high MHC class II expression and induced decreased tumor burden when inoculated s.c. with Lewis lung carcinoma cells. This effect was dependent on the dectin-1 receptor. Strikingly, patients with non-small cell lung carcinoma that had received WGP treatment for 10-14 d prior to any other treatment had a decreased frequency of CD14(-)HLA-DR(-)CD11b(+)CD33(+) MDSC in the peripheral blood. Overall, these data indicate that WGP may be a potent immune modulator of MDSC suppressive function and differentiation in cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Monocitos/inmunología , Neutrófilos/inmunología , beta-Glucanos/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células Presentadoras de Antígenos/inmunología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Western Blotting , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Diferenciación Celular/inmunología , Separación Celular , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Humanos , Lectinas Tipo C/metabolismo , Neoplasias Pulmonares/inmunología , Prueba de Cultivo Mixto de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Monocitos/citología , Células Mieloides/citología , Células Mieloides/inmunología , Neutrófilos/citología , Reacción en Cadena en Tiempo Real de la Polimerasa , Levaduras , beta-Glucanos/inmunología
9.
J Immunol ; 196(11): 4477-86, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27183592

RESUMEN

Ab maturation as well as memory B and plasma cell differentiation occur primarily in the germinal centers (GCs). Systemic lupus erythematosus (SLE) may develop as a result of enhanced GC activity. Previous studies have shown that the dysregulated STAT3 pathway is linked to lupus pathogenesis. However, the exact role of STAT3 in regulating SLE disease progression has not been fully understood. In this study, we demonstrated that STAT3 signaling in B cells is essential for GC formation and maintenance as well as Ab response. Increased cell apoptosis and downregulated Bcl-xL and Mcl-1 antiapoptotic gene expression were found in STAT3-deficient GC B cells. The follicular helper T cell response positively correlated with GC B cells and was significantly decreased in immunized B cell STAT3-deficient mice. STAT3 deficiency also led to the defect of plasma cell differentiation. Furthermore, STAT3 deficiency in autoreactive B cells resulted in decreased autoantibody production. Results obtained from B cell STAT3-deficient B6.MRL/lpr mice suggest that STAT3 signaling significantly contributes to SLE pathogenesis by regulation of GC reactivity, autoantibody production, and kidney pathology. Our findings provide new insights into the role of STAT3 signaling in the maintenance of GC formation and GC B cell differentiation and identify STAT3 as a novel target for treatment of SLE.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Centro Germinal/citología , Centro Germinal/inmunología , Lupus Eritematoso Sistémico/inmunología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT3/deficiencia
10.
J Immunol ; 195(10): 5055-65, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26453753

RESUMEN

Tumor-associated macrophages (TAM) with an alternatively activated phenotype have been linked to tumor-elicited inflammation, immunosuppression, and resistance to chemotherapies in cancer, thus representing an attractive target for an effective cancer immunotherapy. In this study, we demonstrate that particulate yeast-derived ß-glucan, a natural polysaccharide compound, converts polarized alternatively activated macrophages or immunosuppressive TAM into a classically activated phenotype with potent immunostimulating activity. This process is associated with macrophage metabolic reprograming with enhanced glycolysis, Krebs cycle, and glutamine utilization. In addition, particulate ß-glucan converts immunosuppressive TAM via the C-type lectin receptor dectin-1-induced spleen tyrosine kinase-Card9-Erk pathway. Further in vivo studies show that oral particulate ß-glucan treatment significantly delays tumor growth, which is associated with in vivo TAM phenotype conversion and enhanced effector T cell activation. Mice injected with particulate ß-glucan-treated TAM mixed with tumor cells have significantly reduced tumor burden with less blood vascular vessels compared with those with TAM plus tumor cell injection. In addition, macrophage depletion significantly reduced the therapeutic efficacy of particulate ß-glucan in tumor-bearing mice. These findings have established a new paradigm for macrophage polarization and immunosuppressive TAM conversion and shed light on the action mode of ß-glucan treatment in cancer.


Asunto(s)
Polisacáridos Fúngicos/farmacología , Lectinas Tipo C/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/inmunología , Neoplasias Experimentales/tratamiento farmacológico , Saccharomyces cerevisiae/química , beta-Glucanos/farmacología , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Línea Celular Tumoral , Polisacáridos Fúngicos/química , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , beta-Glucanos/química
11.
J Immunol ; 190(11): 5588-99, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23630363

RESUMEN

Ab therapy against surface Ags on tumor cells has demonstrated significant efficacy for some cancers. However, it is costly and patients frequently develop acquired resistance over time. In cases of Ab therapy resistance, T cell responses have been shown to be essential in controlling disease progression. Thus, vaccination that generates a sustained Ab response as well as a T cell response may be more effective and economical. In this article, we have developed a vaccination strategy by targeting protein Ags to B cells via a CD19 single-chain variable fragment miniAb. Using the tumor-associated Ag her-2/neu extracellular domain, we showed that the coengagement of CD19 and BCR induced full B cell activation to produce a high titer of Abs and enhanced CD4 Th2 response and CD8 T cell activation and differentiation. These Abs competitively inhibited humanized her-2/neu Ab binding and were capable of activating the complement and inhibiting human breast cancer growth in vitro. Therapeutic efficacy was demonstrated in vivo using murine mammary carcinoma models. Furthermore, four different extracellular domains of her-2/neu could be targeted to B cells to generate Abs against particular domains with different antitumor properties. This approach may offer a new avenue for vaccine development with significantly lower cost, which may be of use not only for cancer therapy but also for infectious agents.


Asunto(s)
Antígenos CD19/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos B/inmunología , Vacunas contra el Cáncer/inmunología , Neoplasias/inmunología , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD19/metabolismo , Antígenos de Neoplasias/metabolismo , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Citocinas/biosíntesis , Epítopos/inmunología , Humanos , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Ratones , Ratones Transgénicos , Neoplasias/mortalidad , Neoplasias/patología , Neoplasias/terapia , Unión Proteica/inmunología , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/inmunología , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/metabolismo , Células Th2/inmunología , Trastuzumab , Carga Tumoral/inmunología
12.
J Immunol ; 189(6): 2985-94, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22914051

RESUMEN

Promoting complement (C) activation may enhance immunological mechanisms of anti-tumor Abs for tumor destruction. However, C activation components, such as C5a, trigger inflammation, which can promote tumor growth. We addressed the role of C5a on tumor growth by transfecting both human carcinoma and murine lymphoma with mouse C5a. In vitro growth kinetics of C5a, control vector, or parental cells revealed no significant differences. Tumor-bearing mice with C5a-transfected xenografted tumor cells had significantly less tumor burden as compared with control vector tumors. NK cells and macrophages infiltrated C5a-expressing tumors with significantly greater frequency, whereas vascular endothelial growth factor, arginase, and TNF-α production were significantly less. Tumor-bearing mice with high C5a-producing syngeneic lymphoma cells had significantly accelerated tumor progression with more Gr-1+CD11b+ myeloid cells in the spleen and overall decreased CD4+ and CD8+ T cells in the tumor, tumor-draining lymph nodes, and the spleen. In contrast, tumor-bearing mice with low C5a-producing lymphoma cells had a significantly reduced tumor burden with increased IFN-γ-producing CD4+ and CD8+ T cells in the spleen and tumor-draining lymph nodes. These studies suggest concentration of local C5a within the tumor microenvironment is critical in determining its role in tumor progression.


Asunto(s)
Complemento C5a/fisiología , Linfoma/inmunología , Linfoma/patología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Microambiente Tumoral/inmunología , Animales , Carcinoma/genética , Carcinoma/inmunología , Carcinoma/patología , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/inmunología , Técnicas de Cocultivo , Complemento C5a/genética , Complemento C5a/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Inmunidad Innata/genética , Leucocitos/inmunología , Leucocitos/metabolismo , Leucocitos/patología , Linfoma/genética , Ratones , Ratones SCID , Ratones Transgénicos , Neoplasias Ováricas/genética , Microambiente Tumoral/genética
13.
Blood ; 117(25): 6825-36, 2011 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-21531981

RESUMEN

ß-glucans have been reported to function as a potent adjuvant to stimulate innate and adaptive immune responses. However, ß-glucans from different sources are differential in their structure, conformation, and thus biologic activity. Different preparations of ß-glucans, soluble versus particulate, further complicate their mechanism of action. Here we show that yeast-derived particulate ß-glucan activated dendritic cells (DCs) and macrophages via a C-type lectin receptor dectin-1 pathway. Activated DCs by particulate ß-glucan promoted Th1 and cytotoxic T-lymphocyte priming and differentiation in vitro. Treatment of orally administered yeast-derived particulate ß-glucan elicited potent antitumor immune responses and drastically down-regulated immunosuppressive cells, leading to the delayed tumor progression. Deficiency of the dectin-1 receptor completely abrogated particulate ß-glucan-mediated antitumor effects. In contrast, yeast-derived soluble ß-glucan bound to DCs and macrophages independent of the dectin-1 receptor and did not activate DCs. Soluble ß-glucan alone had no therapeutic effect but significantly augmented antitumor monoclonal antibody-mediated therapeutic efficacy via a complement activation pathway but independent of dectin-1 receptor. These findings reveal the importance of different preparations of ß-glucans in the adjuvant therapy and allow for the rational design of immunotherapeutic protocols usable in clinical trials.


Asunto(s)
Inmunidad Adaptativa , Adyuvantes Inmunológicos/uso terapéutico , Inmunidad Innata , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Saccharomyces cerevisiae/química , beta-Glucanos/inmunología , beta-Glucanos/uso terapéutico , Animales , Línea Celular Tumoral , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Lectinas Tipo C , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/inmunología , Fagocitosis , Saccharomyces cerevisiae/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología , beta-Glucanos/aislamiento & purificación
17.
Bio Protoc ; 13(5)2023 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-36908637

RESUMEN

A rigorous determination of effector contributions of tumor-infiltrating immune cells is critical for identifying targetable molecular mechanisms for the development of novel cancer immunotherapies. A tumor/immune cell-admixture model is an advantageous strategy to study tumor immunology as the fundamental methodology is relatively straightforward, while also being adaptable to scale to address increasingly complex research queries. Ultimately, this method can provide robust experimental information to complement more traditional murine models of tumor immunology. Here, we describe a tumor/macrophage-admixture model using bone marrow-derived macrophages to investigate macrophage-dependent tumorigenesis. Additionally, we provide commentary on potential branch points for optimization with other immune cells, experimental techniques, and cancer types.

18.
JCI Insight ; 8(9)2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36976637

RESUMEN

Several preclinical studies have demonstrated that certain cytotoxic drugs enhance metastasis, but the importance of host responses triggered by chemotherapy in regulating cancer metastasis has not been fully explored. Here, we showed that multidose gemcitabine (GEM) treatment promoted breast cancer lung metastasis in a transgenic spontaneous breast cancer model. GEM treatment significantly increased accumulation of CCR2+ macrophages and monocytes in the lungs of tumor-bearing as well as tumor-free mice. These changes were largely caused by chemotherapy-induced reactive myelopoiesis biased toward monocyte development. Mechanistically, enhanced production of mitochondrial ROS was observed in GEM-treated BM Lin-Sca1+c-Kit+ cells and monocytes. Treatment with the mitochondria targeted antioxidant abrogated GEM-induced hyperdifferentiation of BM progenitors. In addition, GEM treatment induced upregulation of host cell-derived CCL2, and knockout of CCR2 signaling abrogated the pro-metastatic host response induced by chemotherapy. Furthermore, chemotherapy treatment resulted in the upregulation of coagulation factor X (FX) in lung interstitial macrophages. Targeting activated FX (FXa) using FXa inhibitor or F10 gene knockdown reduced the pro-metastatic effect of chemotherapy. Together, these studies suggest a potentially novel mechanism for chemotherapy-induced metastasis via the host response-induced accumulation of monocytes/macrophages and interplay between coagulation and inflammation in the lungs.


Asunto(s)
Factor X , Neoplasias Pulmonares , Ratones , Animales , Mielopoyesis , Macrófagos/patología , Monocitos/patología , Neoplasias Pulmonares/patología , Gemcitabina
19.
iScience ; 26(5): 106630, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37192973

RESUMEN

Natural IL-17-producing γδ T cells (γδT17 cells) are unconventional innate-like T cells that undergo functional programming in the fetal thymus. However, the intrinsic metabolic mechanisms of γδT17 cell development remain undefined. Here, we demonstrate that mTORC2, not mTORC1, selectively controls the functional fate commitment of γδT17 cells through regulating transcription factor c-Maf expression. scRNA-seq data suggest that fetal and adult γδT17 cells predominately utilize mitochondrial metabolism. mTORC2 deficiency results in impaired Drp1-mediated mitochondrial fission and mitochondrial dysfunction characterized by mitochondrial membrane potential (ΔΨm) loss, reduced oxidative phosphorylation (OXPHOS), and subsequent ATP depletion. Treatment with the Drp1 inhibitor Mdivi-1 alleviates imiquimod-induced skin inflammation. Reconstitution of intracellular ATP levels by ATP-encapsulated liposome completely rescues γδT17 defect caused by mTORC2 deficiency, revealing the fundamental role of metabolite ATP in γδT17 development. These results provide an in-depth insight into the intrinsic link between the mitochondrial OXPHOS pathway and γδT17 thymic programming and functional acquisition.

20.
J Immunother Cancer ; 11(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37072351

RESUMEN

BACKGROUND: Pancreatic cancer (PC) is a challenging diagnosis that is yet to benefit from the advancements in immuno-oncologic treatments. Irreversible electroporation (IRE), a non-thermal method of tumor ablation, is used in treatment of select patients with locally-advanced unresectable PC and has potentiated the effect of certain immunotherapies. Yeast-derived particulate ß-glucan induces trained innate immunity and successfully reduces murine PC tumor burden. This study tests the hypothesis that IRE may augment ß-glucan induced trained immunity in the treatment of PC. METHODS: ß-Glucan-trained pancreatic myeloid cells were evaluated ex vivo for trained responses and antitumor function after exposure to ablated and unablated tumor-conditioned media. ß-Glucan and IRE combination therapy was tested in an orthotopic murine PC model in wild-type and Rag-/- mice. Tumor immune phenotypes were assessed by flow cytometry. Effect of oral ß-glucan in the murine pancreas was evaluated and used in combination with IRE to treat PC. The peripheral blood of patients with PC taking oral ß-glucan after IRE was evaluated by mass cytometry. RESULTS: IRE-ablated tumor cells elicited a potent trained response ex vivo and augmented antitumor functionality. In vivo, ß-glucan in combination with IRE reduced local and distant tumor burden prolonging survival in a murine orthotopic PC model. This combination augmented immune cell infiltration to the PC tumor microenvironment and potentiated the trained response from tumor-infiltrating myeloid cells. The antitumor effect of this dual therapy occurred independent of the adaptive immune response. Further, orally administered ß-glucan was identified as an alternative route to induce trained immunity in the murine pancreas and prolonged PC survival in combination with IRE. ß-Glucan in vitro treatment also induced trained immunity in peripheral blood monocytes obtained from patients with treatment-naïve PC. Finally, orally administered ß-glucan was found to significantly alter the innate cell landscape within the peripheral blood of five patients with stage III locally-advanced PC who had undergone IRE. CONCLUSIONS: These data highlight a relevant and novel application of trained immunity within the setting of surgical ablation that may stand to benefit patients with PC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , beta-Glucanos , Ratones , Animales , beta-Glucanos/farmacología , beta-Glucanos/uso terapéutico , Inmunidad Entrenada , Neoplasias Pancreáticas/tratamiento farmacológico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Electroporación/métodos , Microambiente Tumoral , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA