Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
bioRxiv ; 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38659853

RESUMEN

Metastasis is the leading cause of cancer-related mortality. Paneth cells provide stem cell niche factors in homeostatic conditions, but the underlying mechanisms of cancer stem cell niche development are unclear. Here we report that Dickkopf-2 (DKK2) is essential for the generation of cancer cells with Paneth cell properties during colon cancer metastasis. Splenic injection of Dkk2-knockout (KO) cancer organoids into C57BL/6 mice resulted in a significant reduction of liver metastases. Transcriptome analysis showed reduction of Paneth cell markers such as lysozymes in KO organoids. Single cell RNA sequencing analyses of murine metastasized colon cancer cells and patient samples identified the presence of lysozyme positive cells with Paneth cell properties including enhanced glycolysis. Further analyses of transcriptome and chromatin accessibility suggested Hepatocyte nuclear factor 4-alpha (HNF4A) as a downstream target of DKK2. Chromatin immunoprecipitation followed by sequencing analysis revealed that HNF4A binds to the promoter region of Sox9, a well-known transcription factor for Paneth cell differentiation. In the liver metastatic foci, DKK2 knockout rescued HNF4A protein levels followed by reduction of lysozyme positive cancer cells. Taken together, DKK2-mediated reduction of HNF4A protein promotes the generation of lysozyme positive cancer cells with Paneth cell properties in the metastasized colon cancers.

2.
Front Immunol ; 14: 1257046, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37885890

RESUMEN

Background: Platelets are rapidly deployed to infection sites and respond to pathogenic molecules via pattern recognition receptors (TLR, NLRP). Dickkopf1 (DKK1) is a quintessential Wnt antagonist produced by a variety of cell types including platelets, endothelial cells, and is known to modulate pro-inflammatory responses in infectious diseases and cancer. Moreover, DKK1 is critical for forming leukocyte-platelet aggregates and induction of type 2 cell-mediated immune responses. Our previous publication showed activated platelets release DKK1 following Leishmania major recognition. Results: Here we probed the role of the key surface virulence glycoconjugate lipophosphoglycan (LPG), on DKK1 production using null mutants deficient in LPG synthesis (Δlpg1- and Δlpg2-). Leishmania-induced DKK1 production was reduced to control levels in the absence of LPG in both mutants and was restored upon re-expression of the cognate LPG1 or LPG2 genes. Furthermore, the formation of leukocyte-platelet aggregates was dependent on LPG. LPG mediated platelet activation and DKK1 production occurs through TLR1/2. Conclusion: Thus, LPG is a key virulence factor that induces DKK1 production from activated platelets, and the circulating DKK1 promotes Th2 cell polarization. This suggests that LPG-activated platelets can drive innate and adaptive immune responses to Leishmania infection.


Asunto(s)
Leishmania major , Receptor Toll-Like 1/metabolismo , Células Endoteliales , Inmunidad , Activación Plaquetaria
3.
PLoS One ; 17(4): e0267913, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35486639

RESUMEN

Systemic lupus erythematosus is a chronic disease characterized by autoantibodies, renal and cutaneous disease, and immune complex formation. Emerging evidence suggests that aberrant DNA repair is an underlying mechanism of lupus development. We previously showed that the POLBY265C/C mutation, which results in development of an aberrant immune repertoire, leads to lupus-like disease in mice. To address whether the hematopoietic compartment is sufficient for lupus development, we transplanted bone marrow cells from POLBY265C/C and POLB+/+ into wild-type congenic mice. Only mice transplanted with the POLBY265C/C bone marrow develop high levels of antinuclear antibodies and renal disease. In conclusion, we show that the hematopoietic compartment harvested from the POLBY265C/C mice is sufficient for development of autoimmune disease.


Asunto(s)
ADN Polimerasa beta/metabolismo , Lupus Eritematoso Sistémico , Animales , Anticuerpos Antinucleares/genética , Autoanticuerpos/genética , Lupus Eritematoso Sistémico/genética , Ratones , Mutación
4.
Mol Cancer Ther ; 21(3): 440-447, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35027482

RESUMEN

There is a need for prognostic markers to select patients most likely to benefit from antibody-drug conjugate (ADC) therapy. We quantified the relationship between pretreatment PET imaging of glycoprotein nonmetastatic melanoma B (gpNMB) with 89Zr-labeled anti-gpNMB antibody ([89Zr]ZrDFO-CR011) and response to ADC therapy (CDX-011) in triple-negative breast cancer. First, we compared different PET imaging metrics and found that standardized uptake values (SUV) and tumor-to-heart SUV ratios were sufficient to delineate differences in radiotracer uptake in the tumor of four different cell- and patient-derived tumor models and achieved high standardized effect sizes. These tumor models with varying levels of gpNMB expression were imaged with [89Zr]ZrDFO-CR011 followed by treatment with a single bolus injection of CDX-011. The percent change in tumor volume relative to baseline (% CTV) was then correlated with SUVmean of [89Zr]ZrDFO-CR011 uptake in the tumor. All gpNMB-positive tumor models responded to CDX-011 over 6 weeks of treatment, except one patient-derived tumor regrew after 4 weeks of treatment. As expected, the gpNMB-negative tumor increased in volume by 130 ± 59% at endpoint. The magnitude of pretreatment SUV had the strongest inverse correlation with the % CTV at 2-4 weeks after treatment with CDX-011 (Spearman ρ = -0.8). However, pretreatment PET imaging with [89Zr]ZrDFO-CR011 did not inform on which tumor types will regrow over time. Other methods will be needed to predict resistance to treatment.


Asunto(s)
Melanoma , Neoplasias de la Mama Triple Negativas , Glicoproteínas , Humanos , Melanoma/tratamiento farmacológico , Glicoproteínas de Membrana , Tomografía de Emisión de Positrones , Radioisótopos/uso terapéutico , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Circonio/uso terapéutico
5.
J Leukoc Biol ; 111(4): 893-901, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34890067

RESUMEN

The human body encounters various challenges. Tissue repair and regeneration processes are augmented after tissue injury to reinstate tissue homeostasis. The Wnt pathway plays a crucial role in tissue repair since it induces target genes required for cell proliferation and differentiation. Since tissue injury causes inflammatory immune responses, it has become increasingly clear that the Wnt ligands can function as immunomodulators while critical for tissue homeostasis. The Wnt pathway and Wnt ligands have been studied extensively in cancer biology and developmental biology. While the Wnt ligands are being studied actively, how the Wnt antagonists and their regulatory mechanisms can modulate immune responses during chronic pathological inflammation remain elusive. This review summarizes DKK family proteins as immunomodulators, aiming to provide an overarching picture for tissue injury and repair. To this end, we first review the Wnt pathway components and DKK family proteins. Next, we will review DKK family proteins (DKK1, 2, and 3) as a new class of immunomodulatory protein in cancer and other chronic inflammatory diseases. Taken together, DKK family proteins and their immunomodulatory functions in chronic inflammatory disorders provide novel insights to understand immune diseases and make them attractive molecular targets for therapeutic intervention.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular , Neoplasias , Humanos , Factores Inmunológicos , Ligandos , Neoplasias/metabolismo , Vía de Señalización Wnt
6.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34819374

RESUMEN

Cancer cells can develop an immunosuppressive tumor microenvironment to control tumor-infiltrating lymphocytes. The underlying mechanisms still remain unclear. Here, we report that mouse and human colon cancer cells acquire lymphocyte membrane proteins including cellular markers such as CD4 and CD45. We observed cell populations harboring both a tumor-specific marker and CD4 in the tumor microenvironment. Sorted cells from these populations were capable of forming organoids, identifying them as cancer cells. Live imaging analysis revealed that lymphocyte membrane proteins were transferred to cancer cells via trogocytosis. As a result of the transfer in vivo, cancer cells also acquired immune regulatory surface proteins such as CTLA4 and Tim3, which suppress activation of immune cells [T. L. Walunas et al, Immunity 1, 405-413 (1994) and L. Monney et al., Nature 415, 536-541 (2002)]. RNA sequencing analysis of ex vivo-cocultured splenocytes with trogocytic cancer cells showed reductions in Th1 activation and natural killer cell signaling pathways compared with the nontrogocytic control. Cancer cell trogocytosis was confirmed in the patient-derived xenograft models of colorectal cancer and head and neck cancer. These findings suggest that cancer cells utilize membrane proteins expressed in lymphocytes, which in turn contribute to the development of the immunosuppressive tumor microenvironment.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Antígeno CTLA-4/metabolismo , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Linfocitos Infiltrantes de Tumor/citología , Animales , Células CACO-2 , Membrana Celular/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Células Madre Hematopoyéticas/citología , Humanos , Sistema Inmunológico , Inmunosupresores , Células Jurkat , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Organoides/metabolismo , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Trogocitosis , Microambiente Tumoral
7.
DNA Repair (Amst) ; 105: 103152, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34186496

RESUMEN

The Polb gene encodes DNA polymerase beta (Pol ß), a DNA polymerase that functions in base excision repair (BER) and microhomology-mediated end-joining. The Pol ß-Y265C protein exhibits low catalytic activity and fidelity, and is also deficient in microhomology-mediated end-joining. We have previously shown that the PolbY265C/+ and PolbY265C/C mice develop lupus. These mice exhibit high levels of antinuclear antibodies and severe glomerulonephritis. We also demonstrated that the low catalytic activity of the Pol ß-Y265C protein resulted in accumulation of BER intermediates that lead to cell death. Debris released from dying cells in our mice could drive development of lupus. We hypothesized that deletion of the Neil1 and Ogg1 DNA glycosylases that act upstream of Pol ß during BER would result in accumulation of fewer BER intermediates, resulting in less severe lupus. We found that high levels of antinuclear antibodies are present in the sera of PolbY265C/+ mice deleted of Ogg1 and Neil1 DNA glycosylases. However, these mice develop significantly less severe renal disease, most likely due to high levels of IgM in their sera.


Asunto(s)
ADN Glicosilasas/metabolismo , ADN Polimerasa beta/metabolismo , Reparación del ADN , Lupus Eritematoso Sistémico/enzimología , Mutación , Estrés Oxidativo , Animales , ADN/metabolismo , ADN Glicosilasas/genética , ADN Polimerasa beta/genética , Modelos Animales de Enfermedad , Eliminación de Gen , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/metabolismo , Ratones
8.
iScience ; 24(5): 102411, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-33997693

RESUMEN

Enhanced stemness in colorectal cancer has been reported and it contributes to aggressive progression, but the underlying mechanisms remain unclear. Here we report a Wnt ligand, Dickkopf-2 (DKK2) is essential for developing colorectal cancer stemness. Genetic depletion of DKK2 in intestinal epithelial or stem cells reduced tumorigenesis and expression of the stem cell marker genes including LGR5 in a model of colitis-associated cancer. Sequential mutations in APC, KRAS, TP53, and SMAD4 genes in colonic organoids revealed a significant increase of DKK2 expression by APC knockout and further increased by additional KRAS and TP53 mutations. Moreover, DKK2 activates proto-oncogene tyrosine-protein kinse Src followed by increased LGR5 expressing cells in colorectal cancer through degradation of HNF4α1 protein. These findings suggest that DKK2 is required for colonic epithelial cells to enhance LGR5 expression during the progression of colorectal cancer.

9.
Dev Neurobiol ; 81(4): 389-399, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33811750

RESUMEN

Zika virus (ZIKV) can generate a number of neurological dysfunctions in infected humans. Here, we tested the potential of human immune cells to protect against ZIKV infection in genetically humanized MISTRG mice. FACS analysis showed robust reconstitution of the mouse spleen with human T cells. Peripheral ZIKV inoculation resulted in infection within the brains of MISTRG mice. Mice that were reconstituted with human peripheral blood mononuclear cells (PBMC) showed a more rapid lethal response to ZIKV than the control mice lacking these immune cells. Immunocytochemical analysis of T cell markers CD3, CD45, or CD8 showed strong T cell presence in the brain, together with robust infection by ZIKV particularly in the excitatory pyramidal and granule neurons of the hippocampus. Infection was also found in cortex, striatum, the dopamine neurons of the substantia nigra, and other brain loci. Infection was considerably less in other regions such as the septum and hypothalamus. These data support the perspective that, rather than exerting a protective function, T cells may underlie some ZIKV-mediated neuropathology in the brain.


Asunto(s)
Enfermedades del Sistema Nervioso , Infección por el Virus Zika , Virus Zika , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Leucocitos Mononucleares/patología , Ratones , Infección por el Virus Zika/patología
10.
J Immunol ; 203(5): 1095-1103, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31427398

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are immature suppressive cells found in tumors and immunological niches. In this article, we highlight the ability of MDSCs to promote IL-17-producing T cells (Th17) and regulatory T cells in addition to suppressing cytotoxic T cells in different tumor models. These interactions between MDSCs and T cells support tumor growth because IL-17 is tumorigenic in many cancer types and regulatory T cells suppress antitumor T cells. Besides T cells, MDSCs promote regulatory B cells and suppress overall B cell function; however, tumor-evoked regulatory B cells also regulate MDSC function, suggesting cross-regulation between MDSCs and B cells. These multiple functions shed light on how MDSCs dysregulate several arms of host immune response. Moreover, MDSCs promote tumor cell survival and angiogenesis to support tumors. Therefore, the multifunctional feature of MDSCs make them attractive immunotherapeutic targets.


Asunto(s)
Células Supresoras de Origen Mieloide/fisiología , Neoplasias/inmunología , Animales , Linfocitos B/inmunología , Comunicación Celular , Humanos , Inmunoterapia , Interleucina-17/biosíntesis , Neoplasias/patología , Neoplasias/terapia , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
11.
Differentiation ; 108: 33-39, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31221431

RESUMEN

The Wnt signaling pathway plays essential roles in tissue or organ homeostasis by regulating cell proliferation and differentiation. Upon tissue or organ injury, inflammation is coupled with tissue repair and regeneration process. The canonical Wnt signaling transduction pathway is crucial for cell proliferation, cell differentiation, and tissue regeneration. Dickkopf1 (DKK1) is a quintessential Wnt antagonist that inhibits the Wnt-mediated tissue repair process. Recent studies reported increased levels of DKK1 in many diseases such as cancer, infection, and musculoskeletal diseases. In many cases, the role of DKK1 has been identified as a pro-inflammatory ligand and the expression levels are associated with poor disease outcomes. A variety of cell types including platelets, endothelial cells, and cancer cells secrete DKK1 upon stimuli. This puts DKK1 in a unique place to view immune responses from multicellular interactions in tissue injury and repair process. In this review, we discuss recent efforts to address the underlying mechanism regarding the pro-inflammatory role of DKK1 in cancer, bone diseases, and other inflammatory diseases.


Asunto(s)
Enfermedades Óseas/metabolismo , Carcinogénesis/metabolismo , Factores Inmunológicos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Wnt/metabolismo , Animales , Humanos , Vía de Señalización Wnt
12.
Cancer Res ; 79(7): 1587-1599, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30786994

RESUMEN

Myeloid-derived suppressor cells (MDSC) promote colorectal cancer by several mechanisms, including suppression of antitumor T cells and production of tumorigenic factors. We previously showed that an intermediate MDSC subset (I-MDSC) is expanded in an intestinal tumor model (ApcMin/+ mice), but the importance of this subset in promoting tumors is unclear. Here, we show that I-MDSCs are a distinct heterogeneous subset due to differential and reduced expression of the monocytic marker, Ly6C, and granulocytic marker, Ly6G. Besides causing necroptotic cell death, receptor-interacting protein kinase 3 (RIPK3) has an alternate function as a signaling component inducing cytokine synthesis. We evaluated whether RIPK3 regulates inflammatory cytokines in I-MDSCs to assess the nonimmunosuppressive function of I-MDSCs in promoting tumors. Inhibition of RIPK3 with the commercially available small-molecule inhibitor GSK 872 showed that RIPK3-mediated inflammation promoted intestinal tumors in two intestinal tumor models, ApcMin/+ mice and an MC38 transplantable tumor model. Mechanistically, RIPK3 signaling in I-MDSC increased tumor size by expanding IL17-producing T cells in MC38 tumors. Collectively, these data suggest RIPK3 signaling as a potential therapeutic target in colorectal cancer. SIGNIFICANCE: The specific role of RIPK3 in intestinal tumors and MDSC function sheds light on a key inflammatory mechanism driving tumorigenesis and allows for possible therapeutic intervention.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Inflamación/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interleucinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores
13.
Trends Immunol ; 39(10): 830-847, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30213499

RESUMEN

Cell differentiation, proliferation, and death are vital for immune homeostasis. Wnt signaling plays essential roles in processes across species. The roles of Wnt signaling proteins and Wnt ligands have been studied in the past, but the context-dependent mechanisms and functions of these pathways in immune responses remain unclear. Recent findings regarding the role of Wnt ligands and Wnt signaling in immune cells and their immunomodulatory mechanisms suggest that Wnt ligands and signaling are significant in regulating immune responses. We introduce recent key findings and future perspectives on Wnt ligands and their signaling pathways in immune cells as well as the immunological roles and functions of Wnt antagonists.


Asunto(s)
Sistema Inmunológico , Inmunidad Celular , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Diferenciación Celular , Homeostasis , Humanos
14.
Biomaterials ; 183: 128-138, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30165256

RESUMEN

Manipulation of human T cell functioning by delivery of macromolecules such as DNA, RNA, or protein is limited, unless the human T cells have been stimulated or electropermeabilized. To achieve successful adaptation and survival of a grafted organ, the alloreactive T cells that induce graft rejection must be regulated. Corticosteroids, calcineurin inhibitors, and mTOR inhibitors, which are systemic immunosuppressants, are currently used for transplantation, with significant side effects. In this study, we demonstrated that a cell-permeable peptide (CPP), dNP2, could efficiently deliver proteins into human CD4 and CD8 T cells. We confirmed regulatory functioning of the cytoplasmic domain of CTLA-4 conjugated with dNP2 (dNP2-ctCTLA-4) in human T cell activation, proliferation, and chemokine receptor expression. We utilized a human skin allograft system in SCID/beige mice to examine whether dNP2-ctCTLA-4 could inhibit allograft rejection by controlling T cell responses. The grafted skin tissue inflammation, allogeneic T cell infiltration, and blood cytokine level was markedly reduced by dNP2-ctCTLA-4, resulting in successful transplantation. In addition, it also inhibited T cell alloresponses against microvessels formed form Bcl-2-transduced human umbilical vein endothelial cells implanted into Balb/c Rag1-/-/IL-2Rγ-/- double knockout (DKO) mice, assessed as reduced T cell infiltration and granzyme B expression. These results collectively suggest that dNP2 peptide conjugation offers a valuable tool for delivering macromolecules like proteins into human T cells, and dNP2-ctCTLA-4 is a novel agent that shows potential in controlling human T cell responses to allow successful adaptation of grafted tissues.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/química , Péptidos de Penetración Celular/química , Rechazo de Injerto/prevención & control , Microvasos/trasplante , Trasplante de Piel , Linfocitos T/efectos de los fármacos , Animales , Antígeno CTLA-4/metabolismo , Proliferación Celular/efectos de los fármacos , Péptidos de Penetración Celular/metabolismo , Citocinas/sangre , Células Endoteliales , Femenino , Rechazo de Injerto/inmunología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/trasplante , Humanos , Activación de Linfocitos , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Receptores de Quimiocina/metabolismo , Piel/inmunología , Linfocitos T/inmunología
15.
Front Immunol ; 9: 303, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29503652

RESUMEN

Regulatory T cells (Tregs) are an important subset of adaptive immune cells and control immune reactions for maintaining homeostasis. Tregs are generated upon their encounter with self or non-self-antigen and mediate tolerance or suppress aberrant immune responses. A high level of specificity of Tregs to recognize antigen(s) suggested their instrumental potential to treat various inflammatory diseases. This review will first introduce seminal basic research findings in the field of Tregs over the last two decades pertinent to therapeutic approaches in progress. We will then discuss the previous approaches to use Tregs for therapeutic purposes and the more recent development of gene-modification approaches. The suppressive function of Tregs has been studied intensively in clinical settings, including cancer, autoimmunity, and allotransplantation. In cancer, Tregs are often aberrantly increased in their number, and their suppressor function inhibits mounting of effective antitumor immune responses. We will examine potential approaches of using gene-modified Tregs to treat cancer. In autoimmunity and allotransplantation, chronic inflammation due to inherent genetic defects in the immune system or mismatch between organ donor and recipient results in dysfunction of Tregs, leading to inflammatory diseases or rejection, respectively. Since the recognition of antigen is a central part in Treg function and their therapeutic use, the modulation of T cell receptor specificity will be discussed. Finally, we will focus on future novel strategies employing the therapeutic potential of Tregs using gene modification to broaden our perspective.


Asunto(s)
Traslado Adoptivo , Enfermedades Autoinmunes , Rechazo de Injerto , Neoplasias , Receptores de Antígenos de Linfocitos T , Linfocitos T Reguladores , Trasplantes , Aloinjertos , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Rechazo de Injerto/terapia , Humanos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante
16.
Nat Med ; 24(3): 262-270, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29431745

RESUMEN

Immunotherapy offers new options for cancer treatment, but efficacy varies across cancer types. Colorectal cancers (CRCs) are largely refractory to immune-checkpoint blockade, which suggests the presence of yet uncharacterized immune-suppressive mechanisms. Here we report that the loss of adenomatosis polyposis coli (APC) in intestinal tumor cells or of the tumor suppressor PTEN in melanoma cells upregulates the expression of Dickkopf-related protein 2 (DKK2), which, together with its receptor LRP5, provides an unconventional mechanism for tumor immune evasion. DKK2 secreted by tumor cells acts on cytotoxic lymphocytes, inhibiting STAT5 signaling by impeding STAT5 nuclear localization via LRP5, but independently of LRP6 and the Wnt-ß-catenin pathway. Genetic or antibody-mediated ablation of DKK2 activates natural killer (NK) cells and CD8+ T cells in tumors, impedes tumor progression, and enhances the effects of PD-1 blockade. Thus, we have identified a previously unknown tumor immune-suppressive mechanism and immunotherapeutic targets particularly relevant for CRCs and a subset of melanomas.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Neoplasias Colorrectales/inmunología , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Melanoma/inmunología , Escape del Tumor/genética , Proteína de la Poliposis Adenomatosa del Colon/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Citotoxicidad Inmunológica/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/inmunología , Neoplasias Intestinales/genética , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/terapia , Células Asesinas Naturales/inmunología , Melanoma/genética , Melanoma/patología , Melanoma/terapia , Fosfohidrolasa PTEN , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Factor de Transcripción STAT5/genética , Transducción de Señal , beta Catenina/genética
17.
Kidney Int ; 93(5): 1118-1130, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29409726

RESUMEN

Excessive expression of Tbet and IFNγ is evidence of systemic lupus erythematosus (SLE) in lupus patients. In this study, the nucleus-transducible form of Transcription Modulation Domain (TMD) of Tbet (ntTbet-TMD), which is a fusion protein between Protein Transduction Domain Hph-1 (Hph-1-PTD) and the TMD of Tbet comprising DNA binding domain and isotype-specific domain, was generated to inhibit Tbet-mediated transcription in the interactomic manner. ntTbet-TMD was effectively delivered into the nucleus of the cells and specifically inhibited Tbet-mediated transcription without influencing the differentiation of other T cell subsets and signaling events for T cell activation. The severity of nephritis was significantly reduced by ntTbet-TMD as effectively as methylprednisolone in lupus-prone mice. The number of Th1, Th2 or Th17 cells and the secretion of their cytokines substantially decreased in the spleen and kidney of lupus-prone mice by ntTbet-TMD treatment. In contrast to methylprednisolone, the marked increase of Treg cells and the secretion of their immunosuppressive cytokine were detected in the spleen of (NZB/NZW) F1 mice treated with ntTbet-TMD. Thus, ntTbet-TMD can improve nephritis in lupus-prone mice by modulating the overall proinflammatory microenvironment and rebalancing T cell subsets, leading to new immune therapeutics for Th1-mediated autoimmune diseases.


Asunto(s)
Antiinflamatorios/farmacología , Núcleo Celular/efectos de los fármacos , Riñón/efectos de los fármacos , Nefritis Lúpica/tratamiento farmacológico , Proteínas de Dominio T Box/farmacología , Transcripción Genética/efectos de los fármacos , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Núcleo Celular/patología , Microambiente Celular , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Nefritis Lúpica/genética , Nefritis Lúpica/inmunología , Nefritis Lúpica/metabolismo , Ratones Endogámicos NZB , Dominios Proteicos , Proteínas Recombinantes/farmacología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
18.
Neoplasia ; 19(8): 595-605, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28654863

RESUMEN

Intestinal tumorigenesis in the ApcMin/+ model is initiated by aberrant activation of Wnt pathway. Increased IL-4 expression in human colorectal cancer tissue and growth of colon cancer cell lines implied that IL-4-induced Stat6-mediated tumorigenic signaling likely contributes to intestinal tumor progression in ApcMin/+ mice. Stat6 also appears to promote expansion of myeloid-derived suppressor cells (MDSCs) cells. MDSCs promote polyp formation in the ApcMin/+ model. Hence, Stat6 could have a broad role in coordinating both polyp cell proliferation and MDSC expansion. We found that IL-4-induced Stat6-mediated proliferation of intestinal epithelial cells is augmented by platelet-derived growth factor-BB, a tumor-promoting growth factor. To determine whether polyp progression in ApcMin/+ mice is dependent on Stat6 signaling, we disrupted Stat6 in this model. Total polyps in the small intestine were fewer in ApcMin/+ mice lacking Stat6. Furthermore, proliferation of polyp epithelial cells was reduced, indicating that Stat6 in part controlled polyp formation. Stat6 also promoted expansion of MDSCs in the spleen and lamina propria of ApcMin/+ mice, implying regulation of antitumor T-cell response. More CD8 cells and reduced PD-1 expression on CD4 cells correlated with reduced polyps. In addition, a strong CD8-mediated cytotoxic response led to killing of tumor cells in Stat6-deficient ApcMin/+ mice. Therefore, these findings show that Stat6 has an oncogenic role in intestinal tumorigenesis by promoting polyp cell proliferation and immunosuppressive mediators, and preventing an active cytotoxic process.


Asunto(s)
Poliposis Adenomatosa del Colon/etiología , Poliposis Adenomatosa del Colon/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Factor de Transcripción STAT6/metabolismo , Poliposis Adenomatosa del Colon/patología , Animales , Becaplermina , Biomarcadores , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Transformación Celular Neoplásica/genética , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Eliminación de Gen , Expresión Génica , Interleucina-4/metabolismo , Interleucina-4/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Intestino Delgado/patología , Ratones , Ratones Noqueados , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Factor de Transcripción STAT6/genética
19.
Immunology ; 152(2): 265-275, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28556921

RESUMEN

Induction of tolerance is a key mechanism to maintain or to restore immunological homeostasis. Here we show that Foxp3+ regulatory T (Treg) cells use Dickkopf-1 (DKK-1) to regulate T-cell-mediated tolerance in the T-cell-mediated autoimmune colitis model. Treg cells from DKK-1 hypomorphic doubleridge mice failed to control CD4+ T-cell proliferation, resulting in CD4 T-cell-mediated autoimmune colitis. Thymus-derived Treg cells showed a robust expression of DKK-1 but not in naive or effector CD4 T cells. DKK-1 expression in Foxp3+ Treg cells was further increased upon T-cell receptor stimulation in vitro and in vivo. Interestingly, Foxp3+ Treg cells expressed DKK-1 in the cell membrane and the functional inhibition of DKK-1 using DKK-1 monoclonal antibody abrogated the suppressor function of Foxp3+ Treg cells. DKK-1 expression was dependent on de novo protein synthesis and regulated by the mitogen-activated protein kinase pathway but not by the canonical Wnt pathway. Taken together, our results highlight membrane-bound DKK-1 as a novel Treg-derived mediator to maintain immunological tolerance in T-cell-mediated autoimmune colitis.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Membrana Celular/metabolismo , Colitis/metabolismo , Colon/metabolismo , Factores de Transcripción Forkhead/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Autotolerancia , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Autoinmunidad , Células CHO , Membrana Celular/inmunología , Proliferación Celular , Colitis/genética , Colitis/inmunología , Colitis/patología , Colon/inmunología , Colon/patología , Cricetulus , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Predisposición Genética a la Enfermedad , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Activación de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fenotipo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Factores de Tiempo , Transfección
20.
J Thorac Dis ; 8(9): E1010-E1012, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27747048
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...