Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nat Immunol ; 21(11): 1444-1455, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32958928

RESUMEN

Acquisition of a lipid-laden phenotype by immune cells has been defined in infectious diseases and atherosclerosis but remains largely uncharacterized in cancer. Here, in breast cancer models, we found that neutrophils are induced to accumulate neutral lipids upon interaction with resident mesenchymal cells in the premetastatic lung. Lung mesenchymal cells elicit this process through repressing the adipose triglyceride lipase (ATGL) activity in neutrophils in prostaglandin E2-dependent and -independent manners. In vivo, neutrophil-specific deletion of genes encoding ATGL or ATGL inhibitory factors altered neutrophil lipid profiles and breast tumor lung metastasis in mice. Mechanistically, lipids stored in lung neutrophils are transported to metastatic tumor cells through a macropinocytosis-lysosome pathway, endowing tumor cells with augmented survival and proliferative capacities. Pharmacological inhibition of macropinocytosis significantly reduced metastatic colonization by breast tumor cells in vivo. Collectively, our work reveals that neutrophils serve as an energy reservoir to fuel breast cancer lung metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Metabolismo de los Lípidos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Células Madre Mesenquimatosas/metabolismo , Neutrófilos/metabolismo , Animales , Biomarcadores , Proliferación Celular , Progresión de la Enfermedad , Endocitosis , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Metástasis de la Neoplasia , Neutrófilos/ultraestructura
2.
Immunity ; 55(8): 1483-1500.e9, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35908547

RESUMEN

Primary tumors are drivers of pre-metastatic niche formation, but the coordination by the secondary organ toward metastatic dissemination is underappreciated. Here, by single-cell RNA sequencing and immunofluorescence, we identified a population of cyclooxygenase 2 (COX-2)-expressing adventitial fibroblasts that remodeled the lung immune microenvironment. At steady state, fibroblasts in the lungs produced prostaglandin E2 (PGE2), which drove dysfunctional dendritic cells (DCs) and suppressive monocytes. This lung-intrinsic stromal program was propagated by tumor-associated inflammation, particularly the pro-inflammatory cytokine interleukin-1ß, supporting a pre-metastatic niche. Genetic ablation of Ptgs2 (encoding COX-2) in fibroblasts was sufficient to reverse the immune-suppressive phenotypes of lung-resident myeloid cells, resulting in heightened immune activation and diminished lung metastasis in multiple breast cancer models. Moreover, the anti-metastatic activity of DC-based therapy and PD-1 blockade was improved by fibroblast-specific Ptgs2 deletion or dual inhibition of PGE2 receptors EP2 and EP4. Collectively, lung-resident fibroblasts reshape the local immune landscape to facilitate breast cancer metastasis.


Asunto(s)
Neoplasias Pulmonares , Subtipo EP2 de Receptores de Prostaglandina E , Ciclooxigenasa 2/genética , Fibroblastos/patología , Humanos , Pulmón/patología , Neoplasias Pulmonares/patología , Subtipo EP4 de Receptores de Prostaglandina E/genética , Microambiente Tumoral
3.
Stem Cells ; 32(2): 327-37, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24123709

RESUMEN

An imbalance between normal adipogenesis and osteogenesis by mesenchymal stem cells (MSCs) has been shown to be related to various human metabolic diseases, such as obesity and osteoporosis; however, the underlying mechanisms remain elusive. We found that the interaction between osteopontin (OPN), an arginine-glycine-aspartate-containing glycoprotein, and integrin αv/ß1 plays a critical role in the lineage determination of MSCs. Although OPN is a well-established marker during osteogenesis, its role in MSC differentiation is still unknown. Our study reveals that blockade of OPN function promoted robust adipogenic differentiation, while inhibiting osteogenic differentiation. Re-expression of OPN restored a normal balance between adipogenesis and osteogenesis in OPN(-/-) MSCs. Retarded bone formation by OPN(-/-) MSCs was also verified by in vivo implantation with hydroxyapatite-tricalcium phosphate, a bone-forming matrix. The role of extracellular OPN in MSC differentiation was further demonstrated by supplementation and neutralization of OPN. Blocking well-known OPN receptors integrin αv/ß1 but not CD44 also affected MSC differentiation. Further studies revealed that OPN inhibits the C/EBPs signaling pathway through integrin αv/ß1. Consistent with these in vitro results, OPN(-/-) mice had a higher fat to total body weight ratio than did wild-type mice. Therefore, our study demonstrates a novel role for OPN-integrin αv/ß1 in regulating MSC differentiation.


Asunto(s)
Adipogénesis/genética , Osteogénesis/genética , Osteopontina/metabolismo , Receptores de Vitronectina/metabolismo , Adipocitos/citología , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Humanos , Células Madre Mesenquimatosas , Ratones , Osteoblastos/metabolismo , Osteopontina/genética , Mapas de Interacción de Proteínas/genética , Receptores de Vitronectina/genética
4.
Trends Immunol ; 33(3): 136-43, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22227317

RESUMEN

Mesenchymal stem cells (MSCs), also called multipotent mesenchymal stromal cells, exist in almost all tissues and are a key cell source for tissue repair and regeneration. Under pathological conditions, such as tissue injury, these cells are mobilized towards the site of damage. Tissue damage is usually accompanied by proinflammatory factors, produced by both innate and adaptive immune responses, to which MSCs are known to respond. Indeed, recent studies have shown that there are bidirectional interactions between MSCs and inflammatory cells, which determine the outcome of MSC-mediated tissue repair processes. Although many details of these interactions remain to be elucidated, we provide here a synthesis of the current status of this newly emerging and rapidly advancing field.


Asunto(s)
Células Madre Mesenquimatosas/inmunología , Inmunidad Adaptativa , Animales , Enfermedad Crónica , Humanos , Inmunidad Innata , Inflamación/inmunología
5.
J Biol Chem ; 288(16): 11074-9, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23449975

RESUMEN

MSCs possess potent immunosuppressive capacity. We have reported that mouse MSCs inhibit T cell proliferation and function via nitric oxide. This immune regulatory capacity of MSCs is induced by the inflammatory cytokines IFNγ together with either TNFα or IL-1ß. This effect of inflammatory cytokines on MSCs is extraordinary; logarithmic increases in the expression of iNOS and chemokines are often observed. To investigate the molecular mechanisms underlying this robust effect of cytokines, we examined the expression of microRNAs in MSCs before and after cytokine treatment. We found that miR-155 is most significantly up-regulated. Furthermore, our results showed that miR-155 inhibits the immunosuppressive capacity of MSCs by reducing iNOS expression. We further demonstrated that miR-155 targets TAK1-binding protein 2 (TAB2) to regulate iNOS expression. Additionally, knockdown of TAB2 reduced iNOS expression. In summary, our study demonstrated that miR-155 inhibits the immunosuppressive capacity of MSCs by reducing iNOS expression by targeting TAB2. Our data revealed a novel role of miR-155 in regulating the immune modulatory activities of MSCs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Regulación Enzimológica de la Expresión Génica/inmunología , Tolerancia Inmunológica/fisiología , Células Madre Mesenquimatosas/inmunología , MicroARNs/inmunología , Óxido Nítrico Sintasa de Tipo II/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Regulación Enzimológica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
6.
Blood ; 119(20): 4636-44, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-22415751

RESUMEN

Tissue inhibitor of metalloproteinase-3 (TIMP-3) is one of a family of proteins inhibiting matrix metalloproteinases, which has also been identified as a mediator for checking inflammation. Meanwhile, it is well known that inflammation causes the activation of the immune response. However, it is not clear whether TIMP-3 plays a role in the immune system. In the present study, we demonstrated a novel function of TIMP-3 in Th1/Th2 polarization through its influence on the antigen-presenting cells. First, TIMP-3 was found strikingly up-regulated by IL-4 during the differentiation of human dendritic cells via the p38MAPK pathway. Second, the expression of costimulatory molecule-CD86 was repressed by TIMP-3. Besides, the induction of IL-12 in matured dendritic cells was significantly inhibited in a PI3K-dependent manner. Furthermore, dendritic cells matured in the presence of TIMP-3 could stimulate allogeneic naive T helper (Th) cells to display a prominent Th2 polarization. Importantly, in an autoimmune disorder-primary immune thrombocytopenia, TIMP-3 showed a statistically positive correlation with IL-4 and platelet count, but a negative correlation with IFN-γ in patient blood samples. Collectively, these in vitro and in vivo data clearly suggested a novel role of TIMP-3 in Th1/Th2 balance in humans.


Asunto(s)
Polaridad Celular/genética , Células Dendríticas/fisiología , Células TH1/fisiología , Células Th2/fisiología , Inhibidor Tisular de Metaloproteinasa-3/fisiología , Adolescente , Adulto , Estudios de Casos y Controles , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Polaridad Celular/efectos de los fármacos , Polaridad Celular/inmunología , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/fisiología , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/genética , Púrpura Trombocitopénica Idiopática/inmunología , Púrpura Trombocitopénica Idiopática/metabolismo , ARN Interferente Pequeño/farmacología , Células TH1/citología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th2/citología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Inhibidor Tisular de Metaloproteinasa-3/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Adulto Joven
7.
J Control Release ; 363: 435-451, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37717658

RESUMEN

It is time for the story of mitochondria and intracellular communication in multidrug resistant cancer to be rewritten. Herein we characterize the extent and cellular advantages of mitochondrial network fusion in multidrug resistant (MDR) breast cancer and have designed a novel nanomedicine that disrupts mitochondrial network fusion and systematically manipulates organelle fusion and function. Combination Organelle Mitochondrial Endoplasmic reticulum Therapy (COMET) is an innovative translational nanomedicine for treating MDR triple negative breast cancer (TNBC) that has superior safety and equivalent efficacy to the current standard of care (paclitaxel). Our study has demonstrated that the increased mitochondrial networks in MDR TNBC contribute to apoptotic resistance and network fusion is mediated by mitofusin2 (MFN2) on the outer mitochondrial membrane. COMET consists of three components; Mitochondrial Network Disrupting (MiND) nanoparticles (NPs) that are loaded with an anti-MFN2 peptide, tunicamycin, and Bam7. The therapeutic rationale of COMET is to reduce the apoptotic threshold in MDR cells with MiND NPs, followed by inducing the endoplasmic reticulum mediated unfolded protein response (UPR) by stressing MDR cells with tunicamycin, and finally, directly inducing mitochondrial apoptosis with Bam7 which is a specific bcl-2 Bax activator. MiND NPs are PEGylated liposomes with the 21 amino acid (2577.98 MW) anti-MFN2 peptide compartmentalized in the aqueous core. Hypoxia (0.5% oxygen) was used to create MDR derivatives of MDA-MB-231 cells and BT-549 cells. Mitochondrial networks were quantified using 3D analysis of 60× live cell images acquired with a Keyence BZ-X710 microscope and MiND NPs effectively fragmented mitochondrial networks in drug sensitive and MDR TNBC cells. The IC50 values, combination index, and dose reduction index derived from dose response studies demonstrate that MiND NPs decrease the apoptotic threshold of both drug sensitive and MDR TNBC cells and COMET is a synergistic drug combination. Complex V (ATP synthase) extracted from bovine cardiac mitochondria was used to assess the effect of MiND NPs on OXPHOS; both MiND NPs and anti-MFN2 peptide solution significantly decrease the activity of mitochondrial complex V and decrease the capacity of OXPHOS. A BacMam viral vector based fluorescent biosensor was used to quantify the unfolded protein response (UPR) at the level of the endoplasmic reticulum and tunicamycin specifically induces the UPR in drug sensitive and MDR TNBC cells. A caspase 3 colorimetric assay demonstrated that the synergistic triple drug combination of COMET increases the ability of Bam7 to specifically induce apoptosis. Dose limiting toxicity and off target effects are a significant challenge for current chemotherapy regimens including paclitaxel. COMET has significantly lower cytotoxicity than paclitaxel in human embryonic kidney epithelial cells and has the potential to fulfill the clinical need for safer cancer therapeutics. COMET is a promising early stage translational nanomedicine for treating MDR TNBC. Manipulating intracellular communication and organelle fusion is a novel approach to treating MDR cancer. The data from this study has rewritten the story of mitochondria, organelle fusion, and intracellular communication and by targeting this intersection, COMET is an exciting new chapter in cancer therapeutics that could transform the clinical outcome of MDR TNBC.


Asunto(s)
Resistencia a Múltiples Medicamentos , Neoplasias de la Mama Triple Negativas , Animales , Bovinos , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Tunicamicina/metabolismo , Tunicamicina/farmacología , Resistencia a Antineoplásicos , Paclitaxel , Mitocondrias , Apoptosis , Retículo Endoplásmico/metabolismo , Péptidos/farmacología , Combinación de Medicamentos , Línea Celular Tumoral
8.
Sci Immunol ; 8(80): eadd5204, 2023 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-36800412

RESUMEN

Neutrophils, the most abundant innate immune cells, function as crucial regulators of the adaptive immune system in diverse pathological conditions, including metastatic cancer. However, it remains largely unknown whether their immunomodulatory functions are intrinsic or acquired within the pathological tissue environment. Here, using mouse models of metastatic breast cancer in the lungs, we show that, although neutrophils isolated from bone marrow (BM) or blood are minimally immunosuppressive, lung-infiltrating neutrophils are robustly suppressive of both T cells and natural killer (NK) cells. We found that this tissue-specific immunosuppressive capacity of neutrophils exists in the steady state and is reinforced by tumor-associated inflammation. Acquisition of potent immunosuppression activity by lung-infiltrating neutrophils was endowed by the lung-resident stroma, specifically CD140a+ mesenchymal cells (MCs) and largely via prostaglandin-endoperoxide synthase 2 (PTGS2), the rate-limiting enzyme for prostaglandin E2 (PGE2) biosynthesis. MC-specific deletion of Ptgs2 or pharmacological inhibition of PGE2 receptors reversed lung neutrophil-mediated immunosuppression and mitigated lung metastasis of breast cancer in vivo. These lung stroma-targeting strategies substantially improved the therapeutic efficacy of adoptive T cell-based immunotherapy in treating metastatic disease in mice. Collectively, our results reveal that the immunoregulatory effects of neutrophils are induced by tissue-resident stroma and that targeting tissue-specific stromal factors represents an effective approach to boost tissue-resident immunity against metastatic disease.


Asunto(s)
Neoplasias Pulmonares , Neutrófilos , Animales , Ratones , Ciclooxigenasa 2 , Pulmón/patología , Células Asesinas Naturales , Melanoma Cutáneo Maligno
9.
J Immunol ; 184(5): 2321-8, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20130212

RESUMEN

Cell-cell adhesion mediated by ICAM-1 and VCAM-1 is critical for T cell activation and leukocyte recruitment to the inflammation site and, therefore, plays an important role in evoking effective immune responses. However, we found that ICAM-1 and VCAM-1 were critical for mesenchymal stem cell (MSC)-mediated immunosuppression. When MSCs were cocultured with T cells in the presence of T cell Ag receptor activation, they significantly upregulated the adhesive capability of T cells due to the increased expression of ICAM-1 and VCAM-1. By comparing the immunosuppressive effect of MSCs toward various subtypes of T cells and the expression of these adhesion molecules, we found that the greater expression of ICAM-1 and VCAM-1 by MSCs, the greater the immunosuppressive capacity that they exhibited. Furthermore, ICAM-1 and VCAM-1 were found to be inducible by the concomitant presence of IFN-gamma and inflammatory cytokines (TNF-alpha or IL-1). Finally, MSC-mediated immunosuppression was significantly reversed in vitro and in vivo when the adhesion molecules were genetically deleted or functionally blocked, which corroborated the importance of cell-cell contact in immunosuppression by MSCs. Taken together, these findings reveal a novel function of adhesion molecules in immunoregulation by MSCs and provide new insights for the clinical studies of antiadhesion therapies in various immune disorders.


Asunto(s)
Mediadores de Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Femenino , Citometría de Flujo , Terapia de Inmunosupresión , Mediadores de Inflamación/inmunología , Molécula 1 de Adhesión Intercelular/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-1/inmunología , Interleucina-1/metabolismo , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Receptor de Interferón gamma
10.
Front Immunol ; 13: 759188, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35126389

RESUMEN

Intracellular cytokine staining (ICS) is a widely employed ex vivo method for quantitative determination of the activation status of immune cells, most often applied to T cells. ICS test samples are commonly prepared from animal or human tissues as unpurified cell mixtures, and cell-specific cytokine signals are subsequently discriminated by gating strategies using flow cytometry. Here, we show that when ICS samples contain Ly6G+ neutrophils, neutrophils are ex vivo activated by an ICS reagent - phorbol myristate acetate (PMA) - which leads to hydrogen peroxide (H2O2) release and death of cytokine-expressing T cells. This artifact is likely to result in overinterpretation of the degree of T cell suppression, misleading immunological research related to cancer, infection, and inflammation. We accordingly devised easily implementable improvements to the ICS method and propose alternative methods for assessing or confirming cellular cytokine expression.


Asunto(s)
Biomarcadores , Citocinas/metabolismo , Activación de Linfocitos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Artefactos , Neoplasias de la Mama , Línea Celular , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo/métodos , Citometría de Flujo/normas , Humanos , Peróxido de Hidrógeno/metabolismo , Espacio Intracelular , Recuento de Leucocitos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Modelos Biológicos , Neutrófilos/metabolismo , Neutrófilos/patología
11.
Cell Metab ; 34(12): 1960-1976.e9, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36476935

RESUMEN

While the distant organ environment is known to support metastasis of primary tumors, its metabolic roles in this process remain underdetermined. Here, in breast cancer models, we found lung-resident mesenchymal cells (MCs) accumulating neutral lipids at the pre-metastatic stage. This was partially mediated by interleukin-1ß (IL-1ß)-induced hypoxia-inducible lipid droplet-associated (HILPDA) that subsequently represses adipose triglyceride lipase (ATGL) activity in lung MCs. MC-specific ablation of the ATGL or HILPDA genes in mice reinforced and reduced lung metastasis of breast cancer respectively, suggesting a metastasis-promoting effect of lipid-laden MCs. Mechanistically, lipid-laden MCs transported their lipids to tumor cells and natural killer (NK) cells via exosome-like vesicles, leading to heightened tumor cell survival and proliferation and NK cell dysfunction. Blockage of IL-1ß, which was effective singly, improved the efficacy of adoptive NK cell immunotherapy in mitigating lung metastasis. Collectively, lung MCs metabolically regulate tumor cells and anti-tumor immunity to facilitate breast cancer lung metastasis.


Asunto(s)
Células Asesinas Naturales , Neoplasias Pulmonares , Animales , Ratones , Pulmón , Lípidos
12.
Nat Commun ; 13(1): 6752, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36347862

RESUMEN

CD8+ T cells are a major prognostic determinant in solid tumors, including colorectal cancer (CRC). However, understanding how the interplay between different immune cells impacts on clinical outcome is still in its infancy. Here, we describe that the interaction of tumor infiltrating neutrophils expressing high levels of CD15 with CD8+ T effector memory cells (TEM) correlates with tumor progression. Mechanistically, stromal cell-derived factor-1 (CXCL12/SDF-1) promotes the retention of neutrophils within tumors, increasing the crosstalk with CD8+ T cells. As a consequence of the contact-mediated interaction with neutrophils, CD8+ T cells are skewed to produce high levels of GZMK, which in turn decreases E-cadherin on the intestinal epithelium and favors tumor progression. Overall, our results highlight the emergence of GZMKhigh CD8+ TEM in non-metastatic CRC tumors as a hallmark driven by the interaction with neutrophils, which could implement current patient stratification and be targeted by novel therapeutics.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias Colorrectales , Humanos , Neutrófilos , Neoplasias Colorrectales/patología , Linfocitos Infiltrantes de Tumor
13.
Stem Cells ; 28(1): 122-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19890981

RESUMEN

The balance between Th1 and Th2 cells is critical for homeostasis of the immune system. Th1 cells can also regulate hematopoietic progenitor cell homeostasis by production of oncostatin M. Here we show that Th1 cell products, but not those of Th2 cells, caused a rapid expansion of lineage(-)Sca-1(+)C-kit(+) (LSK) cells in vivo and in vitro. Among Th1 cytokines, interferon-gamma (IFNgamma) was found to play a major role in this expansion by activating the expression of Sca-1 in lineage(-)Sca-1(-)C-kit(+) cells. This process was dependent on IFNgammaR1 signaling and the STAT1 pathway. Furthermore, those IFNgamma-induced LSK cells had a higher proliferation potential than control LSK cells. In addition, while the overall production of colony-forming units in bone marrow was decreased after IFNgamma treatment, the sorted LSK cells could give rise to a higher yield of colony-forming units. Finally, the IFNgamma-induced hematopoiesis was biased toward the differentiation of myeloid lineages. Therefore, our findings demonstrated a novel role of IFNgamma in activating hematopoietic progenitor cells and provide a new insight into the clinical application of interferon.


Asunto(s)
Antígenos Ly/metabolismo , Linaje de la Célula , Proliferación Celular , Hematopoyesis , Células Madre Hematopoyéticas/inmunología , Interferón gamma/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Proteínas Recombinantes/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Células TH1/inmunología , Células Th2/inmunología , Receptor de Interferón gamma
14.
Stem Cells ; 27(8): 1954-62, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19544427

RESUMEN

Bone marrow-derived mesenchymal stem cells (MSCs) hold great promise for treating immune disorders because of their immunoregulatory capacity, but the mechanism remains controversial. As we show here, the mechanism of MSC-mediated immunosuppression varies among different species. Immunosuppression by human- or monkey-derived MSCs is mediated by indoleamine 2,3-dioxygenase (IDO), whereas mouse MSCs utilize nitric oxide, under the same culture conditions. When the expression of IDO and inducible nitric oxide synthase (iNOS) were examined in human and mouse MSCs after stimulation with their respective inflammatory cytokines, we found that human MSCs expressed extremely high levels of IDO, and very low levels of iNOS, whereas mouse MSCs expressed abundant iNOS and very little IDO. Immunosuppression by human MSCs was not intrinsic, but was induced by inflammatory cytokines and was chemokine-dependent, as it is in mouse. These findings provide critical information about the immunosuppression of MSCs and for better application of MSCs in treating immune disorders.


Asunto(s)
Células Madre Mesenquimatosas/inmunología , Óxido Nítrico/inmunología , Animales , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Tolerancia Inmunológica , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Macaca mulatta , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Especificidad de la Especie
15.
J Immunol ; 181(5): 3277-84, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18713999

RESUMEN

Apoptotic cells induce immunosuppression through unknown mechanisms. To identify the underlying molecular mediators, we examined how apoptotic cells induce immunoregulation by dendritic cells (DC). We found that administration of DC exposed to apoptotic cells (DC(ap)) strongly inhibited the expansion of lymphocytes in draining lymph nodes in vivo and the subsequent Ag-specific activation of these lymphocytes ex vivo. Unexpectedly, DC(ap) supported T cell activation to a similar extent as normal DC in vitro, leading to proliferation and IL-2 production, except that DC(ap) did not support T cell production of IFN-gamma. Surprisingly, when DC(ap) were cocultured with normal DC, they completely lost their ability to support T cell activation, an effect reversed by anti-IFN-gamma or inhibitors of inducible NO synthase (iNOS). As expected, exposure to apoptotic cells rendered DC(ap) capable of producing much more NO in response to exogenous IFN-gamma than normal DC. Furthermore, DC(ap) from iNOS(-/-) or IFN-gammaR1(-/-) mice were not inhibitory in vitro or in vivo. Therefore, the IFN-gamma-induced production of NO by apoptotic cell-sensitized DC plays a key role in apoptotic cell-mediated immunosuppression.


Asunto(s)
Apoptosis/inmunología , Células Dendríticas/inmunología , Interferón gamma/fisiología , Óxido Nítrico/fisiología , Animales , Técnicas de Cocultivo , Ganglios Linfáticos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Linfocitos T/inmunología
16.
Nat Commun ; 11(1): 4387, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32873795

RESUMEN

The role of neutrophils in solid tumor metastasis remains largely controversial. In preclinical models of solid tumors, both pro-metastatic and anti-metastatic effects of neutrophils have been reported. In this study, using mouse models of breast cancer, we demonstrate that the metastasis-modulating effects of neutrophils are dictated by the status of host natural killer (NK) cells. In NK cell-deficient mice, granulocyte colony-stimulating factor-expanded neutrophils show an inhibitory effect on the metastatic colonization of breast tumor cells in the lung. In contrast, in NK cell-competent mice, neutrophils facilitate metastatic colonization in the same tumor models. In an ex vivo neutrophil-NK cell-tumor cell tri-cell co-culture system, neutrophils are shown to potentially suppress the tumoricidal activity of NK cells, while neutrophils themselves are tumoricidal. Intriguingly, these two modulatory effects by neutrophils are both mediated by reactive oxygen species. Collectively, the absence or presence of NK cells, governs the net tumor-modulatory effects of neutrophils.


Asunto(s)
Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Mamarias Animales/inmunología , Neutropenia/prevención & control , Neutrófilos/inmunología , Animales , Línea Celular Tumoral/trasplante , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Humanos , Inyecciones Intravenosas , Pulmón/citología , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/secundario , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/sangre , Neoplasias Mamarias Animales/complicaciones , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos NOD , Neutropenia/sangre , Neutropenia/etiología , Neutropenia/inmunología , Neutrófilos/efectos de los fármacos , Cultivo Primario de Células
17.
Pharmacol Ther ; 200: 42-54, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30998940

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent tissue stem cells that differentiate into a number of mesodermal tissue types, including osteoblasts, adipocytes, chondrocytes and myofibroblasts. MSCs were originally identified in the bone marrow (BM) of humans and other mammals, but recent studies have shown that they are multilineage progenitors in various adult organs and tissues. MSCs that localize at perivascular sites function to rapidly respond to external stimuli and coordinate with the vascular and immune systems to accomplish the wound healing process. Cancer, considered as wounds that never heal, is also accompanied by changes in MSCs that parallels the wound healing response. MSCs are now recognized as key players at distinct steps of tumorigenesis. In this review, we provide an overview of the function of MSCs in wound healing and cancer progression with the goal of providing insight into the development of novel MSC-manipulating strategies for clinical cancer treatment.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Neoplasias , Regeneración , Animales , Carcinogénesis , Humanos , Cicatrización de Heridas
18.
Biochem Biophys Res Commun ; 375(4): 666-70, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18755151

RESUMEN

Bone marrow stromal cells (BMSCs) have been shown to promote the growth and survival of a wide variety of tumors. However, in the present study, we found that BMSCs induced apoptosis of lymphoma cells in the presence of INFgamma and TNF. IFNgamma and TNF dramatically induced the expression of inducible nitric oxide synthase (iNOS) by BMSCs in culture, and BMSCs generated from iNOS knockout mice did not induce apoptosis of lymphoma cells in the presence of IFNgamma and TNF. In addition, we found that IFNgamma and TNF also increased IL-6 expression by BMSCs, and anti-IL-6 further increased the killing of tumor cells by BMSCs. Taken together, our findings indicate that BMSCs induce apoptosis of lymphoma cells in the presence of IFNgamma and TNF, and that the proapoptotic effect of BMSCs is mediated by nitric oxide. Our findings suggest a possibility to harness this proapoptotic feature of BMSCs for the development of novel therapeutic strategy to eliminate tumor cells, especially tumor cells in bone marrow.


Asunto(s)
Apoptosis , Células de la Médula Ósea/efectos de los fármacos , Interferón gamma/farmacología , Linfoma/terapia , Óxido Nítrico/biosíntesis , Factor de Necrosis Tumoral alfa/farmacología , Animales , Células de la Médula Ósea/enzimología , Línea Celular Tumoral , Linfoma/patología , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología
19.
Methods Mol Biol ; 1784: 243-258, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29761404

RESUMEN

Macrophages are a heterogeneous population of innate immune cells and are distributed in most adult tissues. Certain tissue-resident macrophages with a prenatal origin, together with postnatal monocyte-derived macrophages, serve as the host scavenger system to eliminate invading pathogens, malignant cells, senescent cells, dead cells, cellular debris, and other foreign substances. As a key member of the mononuclear phagocyte system, macrophages play essential roles in regulation of prenatal development, tissue homeostasis, and disease progression. Over the past two decades, considerable efforts have been made to generate genetic models of macrophage ablation in mice. These models support investigations of the precise functions of tissue-specific macrophages under physiological and pathological conditions. Herein, we overview the currently available mouse strains for in vivo genetic ablation of macrophages and discuss their respective advantages and limitations.


Asunto(s)
Macrófagos/citología , Modelos Genéticos , Fagocitos/citología , Animales , Linaje de la Célula/genética , Modelos Animales de Enfermedad , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Especificidad de Órganos/genética , Fagocitos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética
20.
Methods Mol Biol ; 1784: 263-275, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29761406

RESUMEN

The Cre/loxP system is a widely applied technology for site-specific genetic manipulation in mice. This system allows for deletion of the genes of interest in specific cells, tissues, and whole organism to generate a diversity of conditional knockout mouse strains. Additionally, the Cre/loxP system is useful for development of cell- and tissue-specific reporter mice for lineage tracing, and cell-specific conditional depletion models in mice. Recently, the Cre/loxP technique was extensively adopted to characterize the monocyte/macrophage biology in mouse models. Compared to other relatively homogenous immune cell types such as neutrophils, mast cells, and basophils, monocytes/macrophages represent a highly heterogeneous population which lack specific markers or transcriptional factors. Though great efforts have been made toward establishing macrophage-specific Cre driver mice in the past decade, all of the current available strains are not perfect with regard to their depletion efficiency and targeting specificity for endogenous macrophages. Here we overview the commonly used Cre driver mouse strains targeting macrophages and discuss their major applications and limitations.


Asunto(s)
Integrasas/genética , Macrófagos/metabolismo , Ratones Noqueados/genética , Biología Molecular/métodos , Animales , Macrófagos/citología , Ratones , Monocitos/citología , Monocitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA