Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L872-L884, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34523355

RESUMEN

The influenza virus infection poses a serious health threat worldwide. Myeloid cells play pivotal roles in regulating innate and adaptive immune defense. A disintegrin and metalloproteinase (ADAM) family of proteins contributes to various immune responses; however, the role of a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) in influenza virus infection remains largely unknown. Herein, we investigated its role, focusing on myeloid cells, during influenza virus infection in mice. ADAM10 gene (Adam10)flox/flox/Lyz2-Cre (Adam10ΔLyz2) and control Adam10flox/flox mice were intranasally infected with 200 plaque-forming units of influenza virus A/H1N1/PR8/34. Adam10ΔLyz2 mice exhibited a significantly higher mortality rate, stronger lung inflammation, and a higher virus titer in the lungs than control mice. Macrophages and inflammatory cytokines, such as TNF-α, IL-1ß, and CCL2, were increased in bronchoalveolar lavage fluid from Adam10ΔLyz2 mice following infection. CD11b+Ly6G-F4/80+ myeloid cells, which had an inflammatory monocyte/macrophage-like phenotype, were significantly increased in the lungs of Adam10ΔLyz2 mice. Adoptive transfer experiments suggested that these cells likely contributed to the poorer prognosis in Adam10ΔLyz2 mice. Seven days after infection, CD11b+Ly6G-F4/80+ lung cells exhibited significantly higher arginase-1 expression levels in Adam10ΔLyz2 mice than in control mice, whereas an arginase-1 inhibitor improved the prognosis of Adam10ΔLyz2 mice. Enhanced granulocyte-macrophage colony-stimulating factor (GM-CSF)/GM-CSF receptor signaling likely contributed to this process. Collectively, these results indicate that myeloid ADAM10 protects against influenza virus pneumonia and may be a promising therapeutic target.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Arginasa/biosíntesis , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Células Mieloides/inmunología , Infecciones por Orthomyxoviridae/patología , Proteína ADAM10/genética , Traslado Adoptivo/métodos , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Arginasa/antagonistas & inhibidores , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Citocinas/análisis , Inmunidad Innata/inmunología , Macrófagos/trasplante , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/trasplante , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/prevención & control , Pronóstico , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
3.
Int J Mol Sci ; 22(4)2021 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-33669419

RESUMEN

We have established an immune cell therapy with immortalized induced pluripotent stem-cell-derived myeloid lines (iPS-ML). The benefits of using iPS-ML are the infinite proliferative capacity and ease of genetic modification. In this study, we introduced 4-1BBL gene to iPS-ML (iPS-ML-41BBL). The analysis of the cell-surface molecules showed that the expression of CD86 was upregulated in iPS-ML-41BBL more than that in control iPS-ML. Cytokine array analysis was performed using supernatants of the spleen cells that were cocultured with iPS-ML or iPS-ML-41BBL. Multiple cytokines that are beneficial to cancer immunotherapy were upregulated. Peritoneal injections of iPS-ML-41BBL inhibited tumor growth of peritoneally disseminated mouse melanoma and prolonged survival of mice compared to that of iPS-ML. Furthermore, the numbers of antigen-specific CD8+ T cells were significantly increased in the spleen and tumor tissues treated with epitope peptide-pulsed iPS-ML-41BBL compared to those treated with control iPS-ML. The number of CXCR6-positive T cells were increased in the tumor tissues after treatment with iPS-ML-41BBL compared to that with control iPS-ML. These results suggest that iPS-ML-41BBL could activate antigen-specific T cells and promote their infiltration into the tumor tissues. Thus, iPS-ML-41BBL may be a candidate for future immune cell therapy aiming to change immunological "cold tumor" to "hot tumor".


Asunto(s)
Ligando 4-1BB/metabolismo , Linfocitos T CD8-positivos/inmunología , Inmunoterapia/métodos , Células Madre Pluripotentes Inducidas/citología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/terapia , Células Mieloides/metabolismo , Células Mieloides/trasplante , Neoplasias Cutáneas/terapia , Animales , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores CXCR6/metabolismo , Neoplasias Cutáneas/patología , Resultado del Tratamiento
4.
Front Immunol ; 12: 625998, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717141

RESUMEN

Kidney transplantation is a primary therapy for end-stage renal disease (ESRD) all the time. But it does not mean that we have fully unraveling the mystery of kidney transplantation and confer every patient favorable prognosis. Immune rejection has always been a stumbling block when we try to increase the success rate of kidney transplantation and improve long-term outcomes. Even if the immune rejection is effectively controlled in acute phase, there is a high possibility that the immune response mediated by chronically activated antibodies will trigger chronic rejection and ultimately lead to graft failure. At present, immunosuppressive agent prepared chemically is mainly used to prevent acute or chronic rejection, but it failed to increase the long-term survival rate of allografts or reduce the incidence of chronic rejection after acute rejection, and is accompanied by many adverse reactions. Therefore, many studies have begun to use immune cells to regulate the immune response in order to control allograft rejection. This article will focus on the latest study and prospects of more popular regulatory myeloid cells in the direction of renal transplantation immunotherapy and introduce their respective progress from experimental research to clinical research.


Asunto(s)
Traslado Adoptivo , Rechazo de Injerto/terapia , Supervivencia de Injerto , Trasplante de Riñón/efectos adversos , Trasplante de Células Madre Mesenquimatosas , Células Mieloides/trasplante , Traslado Adoptivo/efectos adversos , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/trasplante , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Humanos , Inmunosupresores/uso terapéutico , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/trasplante , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Resultado del Tratamiento
5.
Cell Immunol ; 362: 104300, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33582607

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells, which have been characterized for their immunosuppressive capacity through multiple mechanisms. These cells have been extensively studied in the field of tumor immunity. Emerging evidence has highlighted its essential role in maintaining immune tolerance in transplantation and autoimmunity. Because of their robust immune inhibitory activities, there has been growing interest in MDSC-based cellular therapy. Various pre-clinical studies have demonstrated that the adoptive transfer of MDCS represented a promising therapeutic strategy for immune-related disorders. In this review, we summarize relevant studies of MDSC-based cell therapy in transplantation and autoimmune diseases and discuss the challenges and future directions for clinical application of MDSC-based cell therapy.


Asunto(s)
Inmunoterapia/métodos , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/trasplante , Traslado Adoptivo/métodos , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Inmunosupresores/farmacología , Células Mieloides/inmunología , Células Mieloides/trasplante , Trasplantes/inmunología
6.
Front Immunol ; 11: 2194, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33133064

RESUMEN

The development of novel approaches to control unwanted immune responses represents an ambitious goal in the management of a number of clinical conditions, including autoimmunity, autoinflammatory diseases, allergies and replacement therapies, in which the T cell response to self or non-harmful antigens threatens the physiological function of tissues and organs. Current treatments for these conditions rely on the use of non-specific immunosuppressive agents and supportive therapies, which may efficiently dampen inflammation and compensate for organ dysfunction, but they require lifelong treatments not devoid of side effects. These limitations induced researchers to undertake the development of definitive and specific solutions to these disorders: the underlying principle of the novel approaches relies on the idea that empowering the tolerogenic arm of the immune system would restore the immune homeostasis and control the disease. Researchers effort resulted in the development of cell-free strategies, including gene vaccination, protein-based approaches and nanoparticles, and an increasing number of clinical trials tested the ability of adoptive transfer of regulatory cells, including T and myeloid cells. Here we will provide an overview of the most promising approaches currently under development, and we will discuss their potential advantages and limitations. The field is teaching us that the success of these strategies depends primarily on our ability to dampen antigen-specific responses without impairing protective immunity, and to manipulate directly or indirectly the immunomodulatory properties of antigen presenting cells, the ultimate in vivo mediators of tolerance.


Asunto(s)
Traslado Adoptivo , Autoantígenos/inmunología , Enfermedades Autoinmunes , Desensibilización Inmunológica , Tolerancia Inmunológica , Células Mieloides , Linfocitos T Reguladores , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Humanos , Células Mieloides/inmunología , Células Mieloides/trasplante , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante
7.
J Investig Dermatol Symp Proc ; 20(1): S16-S21, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33099378

RESUMEN

Alopecia areata (AA), which is defined as an autoimmune hair loss disease, has a serious impact on the quality of life for patients with AA worldwide. In this study, to our knowledge, a previously unreported method of AA induction in C3H mice has been established and validated. Using this method, we showed that dermal injection of 1-3 million of a mixture of skin cells freshly isolated from AA-affected skin induces AA in more than 80% of healthy mice. Contrary to the previous protocol, the induction of AA by this approach does not need any surgical AA skin grafting, cell manipulation, or high number of activated T cells. We also showed that dermal injection of adherent myeloid cells (mainly CD11b+) in healthy mice is as potent as a mixture of none adherent CD3+ T cells and CD19+ B cells in the induction of AA. Interestingly, most of the mice (7 out of 8) that received non-adherent cells developed AA universalis, whereas most of the mice (5 out of 7) that received adherent cells developed patchy AA. Finally, we found a high number of stage-specific embryonic antigen-expressing cells whose expression in monocytes in an inflammatory disease causes the release of inflammatory cytokines, TNF-α and IL-1ß, from these cells in AA-affected skin.


Asunto(s)
Alopecia Areata/metabolismo , Alopecia Areata/patología , Células Mieloides/metabolismo , Células Mieloides/trasplante , Animales , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Antígeno CD11b/metabolismo , Adhesión Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Antígeno Lewis X/metabolismo , Ratones , Ratones Endogámicos C3H , Antígenos Embrionarios Específico de Estadio/metabolismo
8.
J Immunother Cancer ; 8(1)2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31940589

RESUMEN

BACKGROUND: Macrophages and dendritic cells lacking the transcription factor nuclear factor kappa B p50 are skewed toward a proinflammatory phenotype, with increased cytokine expression and enhanced T cell activation; additionally, murine melanoma, fibrosarcoma, colon carcinoma, and glioblastoma grow slower in p50-/- mice. We therefore evaluated the efficacy of p50-negative immature myeloid cells (p50-IMCs) adoptively transferred into tumor-bearing hosts. Immature cells were used to maximize tumor localization, and pretreatment with 5-fluorouracil (5FU) was examined due to its potential to impair marrow production of myeloid cells, to target tumor myeloid cells and to release tumor neoantigens. METHODS: Wild-type (WT)-IMC or p50-IMC were generated by culturing lineage-negative marrow cells from WT or p50-/- mice in media containing thrombopoietin, stem cell factor and Flt3 ligand for 6 days followed by monocyte colony-stimulating factor for 1 day on ultralow attachment plates. Mice inoculated with Hi-Myc prostate cancer (PCa) cells or K-RasG12D pancreatic ductal carcinoma (PDC)-luciferase cells received 5FU followed 5 days later by three doses of 107 immature myeloid cells (IMC) every 3-4 days. RESULTS: PCa cells grew slower in p50-/- mice, and absence of host p50 prolonged the survival of mice inoculated orthotopically with PDC cells. IMC from Cytomegalovirus (CMV)-luciferase mice localized to tumor, nodes, spleen, marrow, and lung. 5FU followed by p50-IMC slowed PCa and PDC tumor growth, ~3-fold on average, in contrast to 5FU followed by WT-IMC, 5FU alone or p50-IMC alone. Slowed tumor growth was evident for 93% of PCa but only 53% of PDC tumors; we therefore focused on PCa for additional IMC analyses. In PCa, p50-IMC matured into F4/80+ macrophages, as well as CD11b+F4/80-CD11c+ conventional dendritic cells (cDCs). In both tumor and draining lymph nodes, p50-IMC generated more macrophages and cDCs than WT-IMC. Activated tumor CD8+ T cells were increased fivefold by p50-IMC compared with WT-IMC, and antibody-mediated CD8+ T cell depletion obviated slower tumor growth induced by 5FU followed by p50-IMC. CONCLUSIONS: 5FU followed by p50-IMC slows the growth of murine prostate and pancreatic carcinoma and depends on CD8+ T cell activation. Deletion of p50 in patient-derived marrow CD34+ cells and subsequent production of IMC for adoptive transfer may contribute to the therapy of these and additional cancers.


Asunto(s)
Carcinoma Ductal Pancreático/terapia , Inmunoterapia Adoptiva/métodos , Células Mieloides/inmunología , Células Mieloides/trasplante , Subunidad p50 de NF-kappa B/deficiencia , Neoplasias Pancreáticas/terapia , Neoplasias de la Próstata/terapia , Animales , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Femenino , Fluorouracilo/farmacología , Masculino , Ratones , Subunidad p50 de NF-kappa B/inmunología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología
9.
Cancer Sci ; 110(10): 3027-3037, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31348591

RESUMEN

We previously established a method to generate myeloid cells with a proliferative capability from pluripotent stem cells and designated them iPS-ML. Human iPS-ML cells share features with physiological macrophages including the capability to infiltrate into cancer tissues. We observed therapeutic effects of human iPS-ML cells expressing interferon ß (iPS-ML/interferon (IFN)-ß) in xenograft cancer models. However, assessment of host immune system-mediated therapeutic and adverse effects of this therapy is impossible by xenograft models. We currently evaluated the therapeutic effects of a mouse equivalent of human iPS-ML/IFN, a mouse embryonic stem (ES) cell-derived myeloid cell line producing IFN (ES-ML/IFN). The ES-MLs producing IFN-ß (ß-ML) and IFN-γ (γ-ML) and originating from E14 ES cells derived from the 129 mouse strain (H-2b ) were generated, and the MHC (H-2Kb , Db , and I-Ab ) genes of the ES-ML/IFN were disrupted using the clustered regularly interspaced short palindromic repeats (CRISPR)/CAS9 method. We used the ES-ML/IFN to treat allogeneic BALB/c mice (H-2d ) transplanted with Colon26 cancer cells. Treatment with ß-ML but not with γ-ML cells repressed the growth of colon cancer in the peritoneal cavity and liver. The transferred ES-ML/IFN infiltrated into cancer tissues and enhanced infiltration of T cells into cancer tissues. ES-ML/IFN therapy increased the number of immune cells in the lymphoid organs. Sensitization of both cancer antigen-specific CD8+ T cells and natural killer (NK) cells were enhanced by the therapy, and CD8+ T cells were essential for the therapeutic effect, implying that donor MHC-deficient ß-ML exhibited a therapeutic effect through the activation of host immune cells derived from allogeneic recipient mice. The results suggested the usefulness of HLA-deficient human iPS-ML/IFN-ß cells for therapy of HLA-mismatched allogeneic cancer patients.


Asunto(s)
Neoplasias del Colon/terapia , Células Madre Embrionarias/citología , Antígenos de Histocompatibilidad/genética , Interferón beta/metabolismo , Células Mieloides/trasplante , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Células Madre Embrionarias/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Asesinas Naturales/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Ratones Endogámicos BALB C , Células Mieloides/citología , Células Mieloides/metabolismo , Trasplante Homólogo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cancer Immunol Res ; 6(8): 930-940, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30021725

RESUMEN

Stimulation of Toll-like receptor 7 (TLR7) activates myeloid cells and boosts the immune response. Previously, we have shown that stimulation of the inhibitory CD200 receptor (CD200R) suppresses TLR7 signaling and that the absence of CD200R signaling leads to a decreased number of papillomas in mice. Here, we investigated the effects of agonistic anti-CD200R on the antitumor activity of a TLR7 agonist (R848) in a syngeneic mouse tumor model. Intratumoral administration of R848 inhibited the growth of the CT26 colon carcinoma and simultaneously decreased CD200R expression in tumor-infiltrating immune cells. The antitumor effects of R848 were potentiated by anti-CD200R. Successfully treated mice were resistant to rechallenge with the same tumor cells. However, the immediate antitumor effects were independent of lymphocytes, because treatment efficacy was similar in wild-type and Rag1tm1Mom mice. Administration of R848, particularly in combination with anti-CD200R, changed the phenotype of intratumoral myeloid cells. The infiltration with immature MHC-II+ macrophages decreased and in parallel monocytes and immature MHC-II- macrophages increased. Combined treatment decreased the expression of the macrophage markers F4/80, CD206, CD86, CD115, and the ability to produce IL1ß, suggesting a shift in the composition of intratumor myeloid cells. Adoptively transferred CD11b+ myeloid cells, isolated from the tumors of mice treated with R848 and anti-CD200R, inhibited tumor outgrowth in recipient mice. We conclude that administration of agonistic anti-CD200R improves the antitumor effects of TLR7 signaling and changes the local tumor microenvironment, which becomes less supportive of tumor progression. Cancer Immunol Res; 6(8); 930-40. ©2018 AACR.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Imidazoles/uso terapéutico , Glicoproteínas de Membrana/inmunología , Receptor Toll-Like 7/inmunología , Microambiente Tumoral/inmunología , Traslado Adoptivo/métodos , Animales , Antígenos CD/metabolismo , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Femenino , Macrófagos/inmunología , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos BALB C , Células Mieloides/trasplante , Transducción de Señal/inmunología , Receptor Toll-Like 7/agonistas
11.
J Pediatric Infect Dis Soc ; 7(suppl_1): S31-S39, 2018 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-29746680

RESUMEN

Chronic granulomatous disease (CGD) is a congenital disorder characterized by recurrent life-threatening bacterial and fungal infections and development of severe inflammation secondary to a congenital defect in 1 of the 5 phagocyte oxidase (phox) subunits of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex. Hematopoietic cell transplant (HCT) is a curative treatment for patients with CGD that provides donor neutrophils with functional NADPH and superoxide anion production. Many characteristics of CGD, including preexisting infection and inflammation and the potential for cure with mixed-donor chimerism, influence the transplant approach and patient outcome. Because of the dangers of short-term death, graft-versus-host disease, and late effects from chemotherapy, HCT historically has been reserved for patients with high-risk disease and a matched donor. However, as advances in CGD and HCT treatments have evolved, recommendations on transplant eligibility also must be amended, but the development of modern guidelines has proven difficult. In this review, we provide an overview of HCT in patients with CGD, including the debate over HCT indications in them, the unique aspects of CGD that can complicate HCT, and a summary of transplant outcomes.


Asunto(s)
Enfermedad Granulomatosa Crónica/terapia , Trasplante de Células Madre Hematopoyéticas , Acondicionamiento Pretrasplante/métodos , Prueba de Histocompatibilidad , Humanos , Células Mieloides/trasplante , NADPH Oxidasas/fisiología , Neutrófilos/fisiología , Neutrófilos/trasplante , Acondicionamiento Pretrasplante/efectos adversos , Trasplante Homólogo
12.
J Neurovirol ; 24(2): 185-191, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28831774

RESUMEN

While the human immunodeficiency virus (HIV) epidemic was initially characterized by a high prevalence of severe and widespread neurological pathologies, the development of better treatments to suppress viremia over years and even decades has mitigated many of the severe neurological pathologies previously observed. Despite effective treatment, mild neurocognitive impairment and premature cognitive aging are observed in HIV-infected individuals, suggesting a changing but ongoing role of HIV infection in the central nervous system (CNS). Although current therapies are effective in suppressing viremia, they are not curative and patients must remain on life-long treatment or risk recrudescence of virus. Important for the development and evaluation of a cure for HIV will be animal models that recapitulate critical aspects of infection in vivo. In the following, we seek to summarize some of the recent developments in humanized mouse models and their usefulness in modeling HIV infection of the CNS and HIV cure strategies.


Asunto(s)
Complejo SIDA Demencia/tratamiento farmacológico , Fármacos Anti-VIH/farmacología , Disfunción Cognitiva/tratamiento farmacológico , Modelos Animales de Enfermedad , VIH-1/efectos de los fármacos , Complejo SIDA Demencia/fisiopatología , Complejo SIDA Demencia/virología , Animales , Terapia Antirretroviral Altamente Activa , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Encéfalo/virología , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/virología , VIH-1/patogenicidad , VIH-1/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/trasplante , Hígado/citología , Hígado/inmunología , Ratones , Ratones SCID , Células Mieloides/trasplante , Células Mieloides/virología , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/trasplante , Timo/citología , Timo/inmunología , Timo/trasplante , Trasplante Heterólogo
13.
J Hepatobiliary Pancreat Sci ; 24(2): 109-119, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28008721

RESUMEN

BACKGROUND: iPS-ML are myeloid lineage cells with a proliferative capacity derived from induced pluripotent stem (iPS) cells. This study aimed to examine therapeutic effect of iPS-ML producing interferon-ß (iPS-ML/IFN-ß) towards primary and metastatic liver cancer and investigate the mechanism of that effect. METHODS: We established a xenograft model of liver metastasis by injecting the spleen of SCID mice with MKN-45 human gastric cancer cells and also a primary liver cancer model by injecting SK-HEP-1 human hepatocellular carcinoma cells into the liver. After cancer lesions were established, iPS-ML/IFN-ß was administered by intraperitoneal injection, and therapeutic effect was evaluated. RESULTS: The i.p. injection of iPS-ML/IFN-ß resulted in a significant retardation of cancer progression and prolonged mouse survival. The infiltration of i.p. administered iPS-ML into tumor lesions located below the liver capsule was observed, suggesting tumor-directed migration and penetration of the liver capsule by iPS-ML. The IFN-ß concentration in the liver was maintained at levels sufficient to exert an anti-cancer effect for at least 3 days post-injection, accounting for the potent therapeutic effect obtained by injection two to three times per week. CONCLUSIONS: This study demonstrates the therapeutic potential of the iPS-ML/IFN-ß in patients with liver cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/terapia , Células Madre Pluripotentes Inducidas/trasplante , Interferón beta/uso terapéutico , Neoplasias Hepáticas/terapia , Células Mieloides/trasplante , Neoplasias Gástricas/terapia , Animales , Carcinoma Hepatocelular/secundario , Tratamiento Basado en Trasplante de Células y Tejidos , Modelos Animales de Enfermedad , Neoplasias Hepáticas/secundario , Ratones SCID , Neoplasias Gástricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Transl Stroke Res ; 7(4): 322-30, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26997513

RESUMEN

Cell-based therapies including bone-marrow derived mononuclear cells (MNCs) are now widely being studied because of their pleotropic effects and promising results to improve recovery after stroke in animal models. Unlike other types of cell therapies, MNCs is a mixture of lymphoid, myeloid, erythroid, and stem cell populations. Which cell population(s) accounts for the beneficial effects of MNCs in stroke recovery is unclear. In this paper, we employed a mouse stroke model with middle cerebral artery occlusion (MCAo), and used positively and negatively sorted autologous MNCs by MACs to determine which fractions of the MNCs contribute to their beneficial effects. We evaluated the benefits of neurofunctional recovery produced by individual cell lineages within MNCs in a long-term observation study up to 28 days after stroke. Mortality and modulation of inflammation were also compared among different sub-populations. We further studied the impact of neurotoxicity posed by activated microglia in the presence of different cell lineages within MNCs. We concluded that myeloid cell lineage and stem cell/progenitors appeared to be important components within MNCs that contribute to improved outcomes after stroke.


Asunto(s)
Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Leucocitos Mononucleares/fisiología , Recuperación de la Función/fisiología , Accidente Cerebrovascular/cirugía , Animales , Antígenos CD/metabolismo , Atrofia/etiología , Atrofia/patología , Células Cultivadas , Técnicas de Cocultivo , Dactinomicina/análogos & derivados , Dactinomicina/toxicidad , Modelos Animales de Enfermedad , Femenino , Inflamación/inducido químicamente , Leucocitos Mononucleares/efectos de los fármacos , Locomoción , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Células Mieloides/fisiología , Células Mieloides/trasplante , Embarazo , Desempeño Psicomotor/fisiología , Recuperación de la Función/efectos de los fármacos
15.
Stem Cells Dev ; 25(7): 530-41, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26879149

RESUMEN

Poor myeloid engraftment remains a barrier to experimental use of humanized mice. Focusing primarily on peripheral blood cells, we compared the engraftment profile of NOD-scid-IL2Rγc(-/-) (NSG) mice with that of NSG mice transgenic for human membrane stem cell factor (hu-mSCF mice), NSG mice transgenic for human interleukin (IL)-3, granulocyte-macrophage-colony stimulating factor (GM-CSF), and stem cell factor (SGM3 mice). hu-mSCF and SGM3 mice showed enhanced engraftment of human leukocytes compared to NSG mice, and this was reflected in the number of human neutrophils and monocytes present in these strains. Importantly, discrete classical, intermediate, and nonclassical monocyte populations were identifiable in the blood of NSG and hu-mSCF mice, while the nonclassical population was absent in the blood of SGM3 mice. Granulocyte-colony stimulating factor (GCSF) treatment increased the number of blood monocytes in NSG and hu-mSCF mice, and neutrophils in NSG and SGM3 mice; however, this effect appeared to be at least partially dependent on the stem cell donor used to engraft the mice. Furthermore, GCSF treatment resulted in a preferential expansion of nonclassical monocytes in both NSG and hu-mSCF mice. Human tubulointerstitial CD11c(+) cells were present in the kidneys of hu-mSCF mice, while monocytes and neutrophils were identified in the liver of all strains. Bone marrow-derived macrophages prepared from NSG mice were most effective at phagocytosing polystyrene beads. In conclusion, hu-mSCF mice provide the best environment for the generation of human myeloid cells, with GCSF treatment further enhancing peripheral blood human monocyte cell numbers in this strain.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Mieloides/trasplante , Transgenes , Animales , Células Cultivadas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interleucina-3/genética , Interleucina-3/metabolismo , Leucocitos/citología , Ratones , Ratones Endogámicos NOD , Células Mieloides/citología , Células Mieloides/efectos de los fármacos , Neutrófilos/citología , Factor de Células Madre/genética , Factor de Células Madre/metabolismo
16.
Int J Cancer ; 138(11): 2688-99, 2016 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26756887

RESUMEN

While myeloid-derived suppressor cells (MDSCs) have been reported to participate in the promotion of angiogenesis and tumor growth, little is known about their presence and function during perioperative period. Here, we demonstrated that human MDSCs expressing CD11b(+), CD33(+) and HLA-DR(-) significantly increased in lung cancer patients after thoracotomy. CD11b(+) CD33(+) HLA-DR(-) MDSCs isolated 24 hr after surgery from lung cancer patients were more efficient in promoting angiogenesis and tumor growth than MDSCs isolated before surgical operation in allograft tumor model. In addition, CD11b(+) CD33(+) HLA-DR(-) MDSCs produced high levels of MMP-9. Using an experimental lung metastasis mouse model, we demonstrated that the numbers of metastases on lung surface and Gr-1(+) CD11b(+) MDSCs at postoperative period were enhanced in proportion to the degree of surgical manipulation. We also examined that syngeneic bone marrow mesenchymal stem cells (BMSCs) significantly inhibited the induction and proliferation of Gr-1(+) CD11b(+) MDSCs and further prevented lung metastasis formation in the mice undergoing laparotomy. Taken together, our results suggest that postoperatively induced MDSCs were qualified with potent proangiogenic and tumor-promotive ability and this cell population should be considered as a target for preventing postoperative tumor metastasis.


Asunto(s)
Proliferación Celular/genética , Neoplasias Pulmonares/terapia , Células Mieloides/trasplante , Neovascularización Patológica/terapia , Animales , Antígeno CD11b/genética , Citometría de Flujo , Antígenos HLA-DR/genética , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/cirugía , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Metástasis de la Neoplasia , Neovascularización Patológica/patología , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Transpl Immunol ; 34: 8-13, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26733333

RESUMEN

PURPOSE: To explore the effects of adoptive transferring sepsis induced myeloid-derived suppressor cells (iMDSCs) in mice corneal, skin, and combined corneal-skin survival. METHODS: Allogeneic full-thickness corneal transplantation, fully mismatched skin transplantation, and corneal-skin combined transplantation (donor C57BL/6 to recipient Balb/c mice) were performed. Sepsis-induced infectious-MDSCs (iMDSCs), were purified from bone marrow of cecal ligated and punctured (CLP) Balb/c mice. Recipient-derived iMDSCs were adoptively transferred into different recipient groups by retro-orbital injection after surgeries. Corneal and skin grafts were examined and photographed routinely for a period of 45days. Histopathology was performed to evaluate corneal-graft inflammation. Bone marrow and/or corneal grafts in each group were harvested from executed recipients on postoperative days 15, 25, 35. Corneal cells and bone marrow cells were stained with CD11b-PE and Gr1-FITC, analyzed by FACS. RESULTS: iMDSCs were able to significantly prolong allograft survival in both corneal and corneal-skin combined transplant groups. A substantial expansion of MDSCs was observed in recipients' bone marrow, particularly in combined groups at an early stage postoperatively, and accordingly the concentration of MDSCs in corneal grafts increased significantly in adoptive transferred groups. CONCLUSIONS: Sepsis-induced MDSCs may suggest a novel cellular therapeutic approach for preventing various types of allograft rejection.


Asunto(s)
Células de la Médula Ósea/inmunología , Trasplante de Córnea , Supervivencia de Injerto/inmunología , Células Mieloides/inmunología , Sepsis/inmunología , Trasplante de Piel , Alotrasplante Compuesto Vascularizado , Traslado Adoptivo , Animales , Femenino , Terapia de Inmunosupresión , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Mieloides/microbiología , Células Mieloides/trasplante
19.
Biochim Biophys Acta ; 1862(3): 323-8, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-26455341

RESUMEN

The pathogenesis of neurological disorders such as multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD) is multifactorial and incompletely understood. The development of therapies for these disorders of the central nervous system (CNS) is thus far very challenging. Neuroinflammation is one of the processes that contribute to the pathogenesis of CNS diseases, and therefore represents an important therapeutic target. Myeloid cells derived from the bone marrow are ideal candidates for cell therapy in the CNS as they are capable of targeting the brain and providing neuroprotective and anti-inflammatory effects. In this review, experimental and clinical evidence for the therapeutic potential of myeloid cells in neurological disorders will be discussed. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.


Asunto(s)
Enfermedad de Alzheimer/terapia , Esclerosis Amiotrófica Lateral/terapia , Esclerosis Múltiple/terapia , Células Mieloides/trasplante , Enfermedad de Alzheimer/patología , Esclerosis Amiotrófica Lateral/patología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Enfermedades del Sistema Nervioso Central/patología , Humanos , Esclerosis Múltiple/patología
20.
Curr Opin Hematol ; 23(1): 72-7, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26554894

RESUMEN

PURPOSE OF REVIEW: The success of allogeneic haematopoietic stem and progenitor cell (HSPC) transplantations remains inconsistent. Umbilical cord blood (UCB) is a promising source of HSPCs for transplantation, but low cell yield hampers its widespread use. Multiple strategies are being developed to manipulate UCB to either increase HSPC content or enhance bone marrow homing upon transfusion. RECENT FINDINGS: Several ex-vivo manipulation protocols have increased engraftment success in recent phase I/II clinical trials. Additionally, by exploiting knowledge of the transcriptome, mature cells were dedifferentiated into induced haematopoietic stem cells capable of self-renewal and reconstitution of haematopoiesis in vivo. SUMMARY: UCB is a more readily available source of allogeneic transplant material compared with bone marrow and mobilized peripheral blood. However, the number of HSPCs in a graft is correlated to the rate and success of engraftment and UCB grafts typically contain 10 times less cells compared with bone marrow or mobilized peripheral blood grafts that contain around 1 × 108 CD34⁺ cells. Recently, research efforts have focused on increasing UCB engrafting cells in addition to the methods to accelerate engraftment or to provide transient protection and support until engraftment succeeds.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Mieloides/trasplante , Técnicas de Cultivo Celular por Lotes , Reprogramación Celular , Ensayos Clínicos como Asunto , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Cultivo Primario de Células , Trasplante Homólogo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...