Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
J Virol ; 97(4): e0010223, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37022164

RESUMEN

Whether and how a local virus infection affects the hematopoietic system in the bone marrow is largely unknown, unlike with systemic infection. In this study, we showed that influenza A virus (IAV) infection leads to demand-adapted monopoiesis in the bone marrow. The beta interferon (IFN-ß) promoter stimulator 1 (IPS-1)-type I IFN-IFN-α receptor 1 (IFNAR1) axis-mediated signaling was found to induce the emergency expansion of the granulocyte-monocyte progenitor (GMP) population and upregulate the expression of the macrophage colony-stimulating factor receptor (M-CSFR) on bipotent GMPs and monocyte progenitors via the signal transducer and activator of transcription 1 (STAT1), leading to a scaled-back proportion of granulocyte progenitors. To further address the influence of demand-adapted monopoiesis on IAV-induced secondary bacterial infection, IAV-infected wild-type (WT) and Stat1-/- mice were challenged with Streptococcus pneumoniae. Compared with WT mice, Stat1-/- mice did not demonstrate demand-adapted monopoiesis, had more infiltrating granulocytes, and were able to effectively eliminate the bacterial infection. IMPORTANCE Our findings show that influenza A virus infection induces type I interferon (IFN)-mediated emergency hematopoiesis to expand the GMP population in the bone marrow. The type I IFN-STAT1 axis was identified as being involved in mediating the viral-infection-driven demand-adapted monopoiesis by upregulating M-CSFR expression in the GMP population. As secondary bacterial infections often manifest during a viral infection and can lead to severe or even fatal clinical complications, we further assessed the impact of the observed monopoiesis on bacterial clearance. Our results suggest that the resulting decrease in the proportion of granulocytes may play a role in diminishing the IAV-infected host's ability to effectively clear secondary bacterial infection. Our findings not only provide a more complete picture of the modulatory functions of type I IFN but also highlight the need for a more comprehensive understanding of potential changes in hematopoiesis during local infections to better inform clinical interventions.


Asunto(s)
Interferón Tipo I , Infecciones por Orthomyxoviridae , Receptor de Factor Estimulante de Colonias de Macrófagos , Factor de Transcripción STAT1 , Regulación hacia Arriba , Animales , Humanos , Ratones , Virus de la Influenza A/inmunología , Interferón Tipo I/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Infecciones por Orthomyxoviridae/inmunología , Hematopoyesis/inmunología , Células Progenitoras de Granulocitos y Macrófagos/inmunología , Streptococcus pneumoniae/inmunología , Infecciones Neumocócicas/inmunología
2.
Dev Comp Immunol ; 116: 103920, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33189746

RESUMEN

Macrophage colony-stimulating factor (MCSF) is an essential growth factor to control the proliferation, differentiation and survival of the macrophage lineage in vertebrates. Sequences of MCSF have been identified in multiple teleost species, however, the functional investigations of MCSF were documented in only a few species. In this study, we examined the biological activity and the immunomodulatory property of a MCSF homologue, PoMCSF, from Japanese flounder (Paralichthys olivaceus). Structural analysis showed that PoMCSF possesses conserved structural characteristics of MCSF proteins, including a signal peptide, a CSF-1 domain, and a transmembrane region closed to the C-terminal. Under normal physiological condition, PoMCSF expression distributes in all the examined tissues, the highest three tissues are blood, muscle, and head kidney. When infected by extracellular and intracellular bacterial pathogens and viral pathogen, the PoMCSF expression patterns vary with different types of microbial pathogens infection and different immune tissues. In vitro experiment showed recombinant PoMCSF promoted the activity of macrophage. In vivo experiment indicated that PoMCSF overexpression boosted the defensive ability of flounder against Edwardsiella piscicida, a severe fish pathogen that infects multiple species of economically important fish, and regulated the expression of multiple immune-related genes. To explore the relationship between PoMCSF and its receptor PoMCSFR, anti-PoMCSFR antibody was prepared and PoMCSFR knockdown was conducted. The neutralization assay showed that when PoMCSFR was neutralized by its antibody, the role of PoMCSF on host defense against E. piscicida was weakened. Knockdown of PoMCSFR impaired the phagocytic capacity of macrophages. Collectively, these findings suggest that PoMCSF plays a crucial role in the immune defense system of Japanese flounder and the effect of PoMCSF is dependent on PoMCSFR. This study provides new insights into the biological activity of MCSF and the relationship between MCSF and MCSFR in teleost.


Asunto(s)
Resistencia a la Enfermedad/inmunología , Proteínas de Peces/inmunología , Peces Planos/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Secuencia de Aminoácidos , Animales , Citocinas/genética , Edwardsiella tarda/patogenicidad , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/veterinaria , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/microbiología , Proteínas de Peces/genética , Regulación de la Expresión Génica , Riñón Cefálico/inmunología , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/inmunología , Fagocitosis , Receptor de Factor Estimulante de Colonias de Macrófagos/genética
3.
J Crohns Colitis ; 14(10): 1436-1445, 2020 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-32271873

RESUMEN

BACKGROUND AND AIM: The mechanisms underlying the formation of intestinal fibrostrictures [FS] in Crohn's disease [CD] are not fully understood, but activation of fibroblasts and excessive collagen deposition are supposed to contribute to the development of FS. Here we investigated whether interleukin-34 [IL-34], a cytokine that is over-produced in CD, regulates collagen production by gut fibroblasts. METHODS: IL-34 and its receptor macrophage colony-stimulating factor receptor 1 [M-CSFR-1] were evaluated in inflammatory [I], FS CD, and control [CTR] ileal mucosal samples by real-time polymerase chain reaction [RT-PCR], western blotting, and immunohistochemistry. IL-34 and M-CSFR-1 expression was evaluated in normal and FS CD fibroblasts. Control fibroblasts were stimulated with IL-34 in the presence or absence of a MAP kinase p38 inhibitor, and FS CD fibroblasts were cultured with a specific IL-34 antisense oligonucleotide, and collagen production was evaluated by RT-PCR, western blotting, and Sircol assay. The effect of IL-34 on the wound healing capacity of fibroblasts was evaluated by scratch test. RESULTS: We showed enhanced M-CSFR-1 and IL-34 RNA and protein expression in FS CD mucosal samples as compared with ICD and CTR samples. Immunohistochemical analysis showed that stromal cells were positive for M-CSFR-1 and IL-34. Enhanced M-CSFR-1 and IL-34 RNA and protein expression was seen in FS CD fibroblasts as compared with CTR. Stimulation of control fibroblasts with IL-34 enhanced COL1A1 and COL3A1 expression and secretion of collagen through a p38 MAP kinase-dependent mechanism, and wound healing. IL-34 knockdown in FS CD fibroblasts was associated with reduced collagen production and wound repair. CONCLUSIONS: Data indicate a prominent role of IL-34 in the control of intestinal fibrogenesis.


Asunto(s)
Colágeno/biosíntesis , Enfermedad de Crohn , Interleucinas/inmunología , Intestinos/patología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Células Cultivadas , Constricción Patológica/etiología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Fibroblastos/metabolismo , Fibrosis/inmunología , Humanos , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Sistema de Señalización de MAP Quinasas/inmunología , Cicatrización de Heridas/inmunología
4.
Dev Comp Immunol ; 98: 89-97, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31029710

RESUMEN

Pathogens such as the Frog Virus 3 (FV3) ranavirus are contributing to the worldwide amphibian declines. While amphibian macrophages (Mϕs) are central to the immune defenses against these viruses, the pathogen recognition capacities of disparate amphibian Mϕ subsets remain unexplored. In turn, Mϕ differentiation and functionality are interdependent on the colony-stimulating factor-1 receptor (CSF-1R), which is ligated by colony-stimulating factor-1 (CSF-1) and the unrelated interleukin-34 (IL-34) cytokines. Notably, the Xenopus laevis frog CSF-1- and IL-34-derived Mϕs are functionally distinct, and while the CSF-1-Mϕs are more susceptible to FV3, the IL-34-Mϕs are highly resistant to this pathogen. Here, we elucidate the pathogen recognition capacities of CSF-1- and IL-34-differentiated Mϕs by evaluating their baseline transcript levels of key pathogen pattern recognition receptors (PRRs). Compared to the frog CSF-1-Mϕs, their IL-34-Mϕs exhibited greater expression of PRR genes associated with viral recognition as well as PRR genes known for recognizing bacterial pathogen-associated molecular patterns (PAMPs). By contrast, the CSF-1-Mϕs displayed greater expression of toll-like receptors (TLRs) that are absent in humans. Moreover, although the two Mϕ types possessed similar expression of most downstream PRR signaling components, they exhibited distinct outcomes upon stimulation with hallmark PAMPs, as measured by their tumor necrosis factor-alpha and interferon-7 gene expression. Remarkably, stimulation with a TLR2/6 agonist conferred FV3 resistance to the otherwise susceptible CSF-1-Mϕs while treatment with a TLR9 agonist significantly ablated the IL-34-Mϕ resistance to FV3. These changes in Mϕ-FV3 susceptibility and resistance appeared to be linked to changes in their expression of key immune genes. Greater understanding of the amphibian macrophage pathogen-recognition capacities will lend to further insights into the pathogen-associated causes of the amphibian declines.


Asunto(s)
Diferenciación Celular/inmunología , Interleucinas/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/inmunología , Ranavirus/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Proteínas de Xenopus/inmunología , Animales , Interacciones Huésped-Patógeno/inmunología , Humanos , Interleucinas/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Macrófagos/virología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Ranavirus/fisiología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Proteínas de Xenopus/metabolismo
5.
J Vis Exp ; (145)2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30933072

RESUMEN

Bone remodeling is a complex process and it involves periods of deposition and resorption. Bone resorption is a process by which bone is broken down by osteoclasts in response to different stimuli. Osteoclast precursors differentiate into multinuclear osteoclasts in response to macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor Kappa-B ligand (RANKL). Under pathologic conditions, the cytokine profile is different and involves a mixture of inflammatory cytokines. Tumor necrosis factor alpha (TNF-α) is one of the most important cytokines as it is found in large amounts in areas involved with inflammatory osteolysis. The purpose of this protocol is to provide a method by which murine bone marrow is isolated to generate osteoclasts through induction with M-CSF and either RANKL or TNF-α which will be subsequently inhibited by increasing doses of anti-c-fms antibody, the receptor for M-CSF. This experiment highlights the therapeutic value of anti-c-fms antibody in diseases of inflammatory bone resorption.


Asunto(s)
Anticuerpos/farmacología , Osteoclastos/citología , Osteoclastos/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Ligando RANK/farmacología , Factor de Necrosis Tumoral alfa/farmacología
6.
J Immunother Cancer ; 6(1): 118, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30424804

RESUMEN

BACKGROUND: The pancreatic cancer vaccine, GVAX, induces novel lymphoid aggregates in the otherwise immune quiescent pancreatic ductal adenocarcinoma (PDAC). GVAX also upregulates the PD-1/PD-L1 pathway, and a pre-clinical model demonstrated the anti-tumor effects of combination GVAX and anti-PD-1 antibody therapy (GVAX/αPD-1). Resistance to GVAX was associated with an immune-suppressive myeloid cell infiltration, which may limit further therapeutic gains of GVAX/αPD-1 therapy. The expression of CSF-1R, a receptor important for myeloid cell migration, differentiation and survival, and the effect of its therapeutic blockade in the context of GVAX in PDAC has not been investigated. METHODS: Lymphoid aggregates appreciated in 24 surgically resected PDAC from patients who received one dose of neoadjuvant GVAX were analyzed with multiplex immunohistochemistry. Flow cytometry analysis of tumor infiltrating T-cells in a murine model of PDAC was performed to investigate the therapeutic effects and mechanism of anti-CSF-1R/anti-PD-1/GVAX combination immunotherapy. RESULTS: High CSF-1R expression in resected PDAC from patients who received neoadjuvant GVAX was associated with a higher myeloid to lymphoid cell ratio (p < 0.05), which has been associated with poorer survival. This higher CSF-1R expression was associated with a higher intra-tumoral infiltration of immature dendritic cells (p < 0.05), but not mature dendritic cells (p = 0.132). In the pre-clinical murine model, administering anti-CSF-1R antibody prior to and after GVAX/αPD-1 ("pre/post-αCSF-1R + αPD-1 + GVAX") enhanced the survival rate compared to GVAX/αPD-1 dual therapy (p = 0.005), but administering anti-CSF-1R only before GVAX/αPD-1 did not (p = 0.41). The "pre/post-αCSF-1R + αPD-1 + GVAX" group also had higher intra-tumoral infiltration of PD-1 + CD8+ and PD-1 + CD4+ T-cells compared to αPD-1/GVAX (p < 0.001). Furthermore, this regimen increased the intra-tumoral infiltration of PD-1 + CD137 + CD8+, PD-1 + CD137 + CD4+ and PD-1 + OX40 + CD4+ T-cells (p < 0.001). These PD-1 + CD137 + CD8+ T-cells expressed high levels of interferon-γ (median 80-90%) in response to stimulation with CD3/CD28 activation beads, and this expression was higher than that of PD-1 + CD137-CD8+ T-cells (p < 0.001). CONCLUSIONS: The conversion of exhausted PD-1+ T-cells to CD137+ activated effector T-cells may contribute to the anti-tumor effects of the anti-CSF-1R/anti-PD-1/GVAX combination therapy. Anti-CSF-1R antibody with anti-PD-1 antibody and GVAX have the potential be an effective therapeutic strategy for treatment of PDAC.


Asunto(s)
Adenocarcinoma/inmunología , Carcinoma Ductal Pancreático/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Tasa de Supervivencia
7.
J Immunol ; 201(7): 2042-2053, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30120124

RESUMEN

Pain is one of the most debilitating symptoms in many diseases for which there is inadequate management and understanding. CSF-1, also known as M-CSF, acts via its receptor (CSF-1R, c-Fms) to regulate the development of the monocyte/macrophage lineage and to act locally in tissues to control macrophage numbers and function. It has been implicated in the control of neuropathic pain via a central action on microglia. We report in this study that systemic administration of a neutralizing anti-CSF-1R or CSF-1 mAb inhibits the development of inflammatory pain induced by zymosan, GM-CSF, and TNF in mice. This approach also prevented but did not ameliorate the development of arthritic pain and optimal disease driven by the three stimuli in mice, suggesting that CSF-1 may only be relevant when the driving inflammatory insults in tissues are acute and/or periodic. Systemic CSF-1 administration rapidly induced pain and enhanced the arthritis in an inflamed mouse joint, albeit via a different pathway(s) from that used by systemic GM-CSF and TNF. It is concluded that CSF-1 can function peripherally during the generation of inflammatory pain and hence may be a target for such pain and associated disease, including when the clinically important cytokines, TNF and GM-CSF, are involved. Our findings have ramifications for the selection and design of anti-CSF-1R/CSF-1 trials.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Inflamación/inmunología , Articulaciones/inmunología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/fisiología , Monocitos/fisiología , Animales , Anticuerpos Neutralizantes/administración & dosificación , Diferenciación Celular , Linaje de la Célula , Humanos , Factor Estimulante de Colonias de Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Dolor , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Transducción de Señal
8.
Proc Natl Acad Sci U S A ; 115(17): E4041-E4050, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29632196

RESUMEN

In a large proportion of cancer patients, CD8 T cells are excluded from the vicinity of cancer cells. The inability of CD8 T cells to reach tumor cells is considered an important mechanism of resistance to cancer immunotherapy. We show that, in human lung squamous-cell carcinomas, exclusion of CD8 T cells from tumor islets is correlated with a poor clinical outcome and with a low lymphocyte motility, as assessed by dynamic imaging on fresh tumor slices. In the tumor stroma, macrophages mediate lymphocyte trapping by forming long-lasting interactions with CD8 T cells. Using a mouse tumor model with well-defined stromal and tumor cell areas, macrophages were depleted with PLX3397, an inhibitor of colony-stimulating factor-1 receptor (CSF-1R). Our results reveal that a CSF-1R blockade enhances CD8 T cell migration and infiltration into tumor islets. Although this treatment alone has minor effects on tumor growth, its combination with anti-PD-1 therapy further increases the accumulation of CD8 T cells in close contact with malignant cells and delays tumor progression. These data suggest that the reduction of macrophage-mediated T cell exclusion increases tumor surveillance by CD8 T cells and renders tumors more responsive to anti-PD-1 treatment.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/inmunología , Macrófagos/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Aminopiridinas/farmacología , Animales , Linfocitos T CD8-positivos/patología , Carcinoma de Células Escamosas/patología , Estudios de Seguimiento , Macrófagos/patología , Ratones , Receptor de Muerte Celular Programada 1/inmunología , Pirroles/farmacología , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Estudios Retrospectivos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Blood ; 131(14): 1587-1599, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29463562

RESUMEN

Monocytes and macrophages play a key role in defending pathogens, removing the dead cells or cell debris, and wound healing. The mammalian target of rapamycin (mTOR) inhibitor rapamycin (RPM) is widely used in clinics to treat patients with organ transplantation or tumors. The role of mTOR in monocyte/macrophage development remains to be clarified. Here we found that mTOR intrinsically controls monocyte/macrophage development, as evidenced by the decreased percentages and cell numbers of CD11b+F4/80+ cells resulting from mTOR inhibition in SCID mice, mTOR-deficient mice, and mixed chimera mice, and the in vitro colony formation and monocyte/macrophage induction assays. However, Lyzs-mTOR knockout mice displayed normal levels of monocytes/macrophages, indicating that mTOR is not essential for the survival and maturation of monocytes/macrophages. Further studies showed that mTOR deficiency significantly reduced macrophage colony-stimulating factor receptor CD115 expression at the transcriptional and translational levels. The molecular mechanism studies indicate that the impaired monocyte/macrophage development caused by mTOR deficiency is mainly a result of the overactivated STAT5 and subsequent downregulation of IRF8, but not the altered cell metabolism and autophagy. Therefore, our work identifies that mTOR is an intrinsic master for monocyte/macrophage development at the early stages through regulating STAT5-IRF8-dependent CD115-expressing pathway. Long-term usage of RPM may cause a defect of myeloid progenitors in bone marrow.


Asunto(s)
Médula Ósea/inmunología , Factores Reguladores del Interferón/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Factor de Transcripción STAT5/inmunología , Serina-Treonina Quinasas TOR/inmunología , Animales , Regulación de la Expresión Génica/inmunología , Humanos , Factores Reguladores del Interferón/genética , Macrófagos/citología , Ratones , Ratones Noqueados , Ratones SCID , Monocitos/citología , Biosíntesis de Proteínas/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Factor de Transcripción STAT5/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/genética , Transcripción Genética/inmunología
10.
J Leukoc Biol ; 103(3): 545-558, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29345363

RESUMEN

Tumor-associated Mφs display a plastic phenotype that is regulated by the local tumor milieu. Gene expression analysis and functional characterization of Mφs exposed in vitro to individual cytokines aids to delineate the cross-talk between defined cytokines shaping the complex Mφ phenotype. Human monocyte-derived Mφs can be differentiated in vitro with the T helper cell type 2 response cytokines IL-4 and IL-13 or the immunosuppressive IL-10. Notably, only the latter subset undergoes apoptosis when treated with the CSF 1 receptor (CSF1R) blocking antibody emactuzumab. However, under physiologic conditions, the Mφ phenotype is regulated by cytokine combination. Hence, in this study, we characterized the plasticity of IL-4 or IL-13-differentiated Mφs upon exposure to the immunosuppressive IL-10. Although IL-4-differentiated Mφs sustained their molecular phenotype in the presence of IL-10, IL-13-differentiated Mφs were skewed towards the IL-10 phenotype. Gene expression profiling revealed unique IL-4+IL-10 and IL-13+IL-10 Mφ signatures associated with up-regulation of canonical NF-κB or Wnt/ß-catenin signaling pathways, respectively. Although IL-10 was able to alter the surface marker and gene expression profile of IL-13-differentiated Mφs, addition of IL-10 did not restore emactuzumab susceptibility. Combining NF-κB and Wnt/ß-catenin signaling inhibitors with emactuzumab had no effect on viability. On average 3-5% of cancer patients overexpressed IL-4, IL-13, or IL-10 mRNA in silico. Although a small patient subset overexpressed IL-10+IL-13, IL-4+IL-10 lacked co-expression. In vitro characterization of CSF1R inhibitor-refractory Mφ phenotypes can support novel pharmacological approaches to specifically target these cells.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Citocinas/farmacología , Interleucina-10/farmacología , Macrófagos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Células Th2/metabolismo , Anticuerpos Monoclonales Humanizados , Células Cultivadas , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Transcriptoma/efectos de los fármacos
11.
J Biol Chem ; 293(5): 1736-1744, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29259133

RESUMEN

Infections by ranaviruses such as Frog virus 3 (Fv3), are significantly contributing to worldwide amphibian population declines. Notably, amphibian macrophages (Mφs) are important to both the Fv3 infection strategies and the immune defense against this pathogen. However, the mechanisms underlying amphibian Mφ Fv3 susceptibility and resistance remain unknown. Mφ differentiation is mediated by signaling through the colony-stimulating factor-1 receptor (CSF-1R) which is now known to be bound not only by CSF-1, but also by the unrelated interleukin-34 (IL-34) cytokine. Pertinently, amphibian (Xenopus laevis) Mφs differentiated by CSF-1 and IL-34 are highly susceptible and resistant to Fv3, respectively. Accordingly, in the present work, we elucidate the facets of this Mφ Fv3 susceptibility and resistance. Because cellular resistance to viral replication is marked by expression of antiviral restriction factors, it was intuitive to find that IL-34-Mφs possess significantly greater mRNA levels of select restriction factor genes than CSF-1-Mφs. Xenopodinae amphibians have highly expanded repertoires of antiviral interferon (IFN) cytokine gene families, and our results indicated that in comparison with the X. laevis CSF-1-Mφs, the IL-34-Mφs express substantially greater transcripts of representative IFN genes, belonging to distinct gene family clades, as well as their cognate receptor genes. Finally, we demonstrate that IL-34-Mφ-conditioned supernatants confer IFN-mediated anti-Fv3 protection to the virally susceptible X. laevis kidney (A6) cell line. Together, this work underlines the differentiation pathways leading to Fv3-susceptible and -resistant amphibian Mφ populations and defines the molecular mechanisms responsible for these differences.


Asunto(s)
Diferenciación Celular/inmunología , Infecciones por Virus ADN/inmunología , Inmunidad Innata , Macrófagos/inmunología , Ranavirus/inmunología , Animales , Interferones/inmunología , Interleucinas/inmunología , Macrófagos/virología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Proteínas de Xenopus/inmunología , Xenopus laevis
12.
Cell Stem Cell ; 21(1): 120-134.e7, 2017 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-28506464

RESUMEN

To investigate the role of immune cells in lung regeneration, we used a unilateral pneumonectomy model that promotes the formation of new alveoli in the remaining lobes. Immunofluorescence and single-cell RNA sequencing found CD115+ and CCR2+ monocytes and M2-like macrophages accumulating in the lung during the peak of type 2 alveolar epithelial stem cell (AEC2) proliferation. Genetic loss of function in mice and adoptive transfer studies revealed that bone marrow-derived macrophages (BMDMs) traffic to the lung through a CCL2-CCR2 chemokine axis and are required for optimal lung regeneration, along with Il4ra-expressing leukocytes. Our data suggest that these cells modulate AEC2 proliferation and differentiation. Finally, we provide evidence that group 2 innate lymphoid cells are a source of IL-13, which promotes lung regeneration. Together, our data highlight the potential for immunomodulatory therapies to stimulate alveologenesis in adults.


Asunto(s)
Pulmón/fisiología , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Neumonectomía , Regeneración/inmunología , Células Th2/inmunología , Animales , Interleucina-13/genética , Interleucina-13/inmunología , Ratones , Ratones Noqueados , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptores CCR2/genética , Receptores CCR2/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Regeneración/genética
13.
Cancer Immunol Res ; 5(7): 535-546, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28536100

RESUMEN

New immunotherapeutic strategies are needed to induce effective antitumor immunity in all cancer patients. Malignant mesothelioma is characterized by a poor prognosis and resistance to conventional therapies. Infiltration of tumor-associated macrophages (TAM) is prominent in mesothelioma and is linked to immune suppression, angiogenesis, and tumor aggressiveness. Therefore, TAM depletion could potentially reactivate antitumor immunity. We show that M-CSFR inhibition using the CSF-1R kinase inhibitor PLX3397 (pexidartinib) effectively reduced numbers of TAMs, circulating nonclassical monocytes, as well as amount of neoangiogenesis and ascites in mesothelioma mouse models, but did not improve survival. When combined with dendritic cell vaccination, survival was synergistically enhanced with a concomitant decrease in TAMs and an increase in CD8+ T-cell numbers and functionality. Total as well as tumor antigen-specific CD8+ T cells in tumor tissue of mice treated with combination therapy showed reduced surface expression of the programmed cell death protein-1 (PD-1), a phenomenon associated with T-cell exhaustion. Finally, mice treated with combination therapy were protected from tumor rechallenge and displayed superior T-cell memory responses. We report that decreasing local TAM-mediated immune suppression without immune activation does not improve survival. However, combination of TAM-mediated immune suppression with dendritic cell immunotherapy generates robust and durable antitumor immunity. These findings provide insights into the interaction between immunotherapy-induced antitumor T cells and TAMs and offer a therapeutic strategy for mesothelioma treatment. Cancer Immunol Res; 5(7); 535-46. ©2017 AACR.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Inmunoterapia Adoptiva , Neoplasias Pulmonares/inmunología , Mesotelioma/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Aminopiridinas/administración & dosificación , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/terapia , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Mesotelioma/inducido químicamente , Mesotelioma/terapia , Mesotelioma Maligno , Invasividad Neoplásica/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirroles/administración & dosificación
14.
Neuro Oncol ; 19(6): 796-807, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28115578

RESUMEN

Background: Adaptive immune resistance in the tumor microenvironment appears to attenuate the immunotherapeutic targeting of glioblastoma (GBM). In this study, we identified a tumor-infiltrating myeloid cell (TIM) population that expands in response to dendritic cell (DC) vaccine treatment. The aim of this study was to understand how this programmed death ligand 1 (PD-L1)-expressing population restricts activation and tumor-cytolytic function of vaccine-induced tumor-infiltrating lymphocytes (TILs). Methods: To test this hypothesis in our in vivo preclinical model, we treated mice bearing intracranial gliomas with DC vaccination ± murine anti-PD-1 monoclonal antibody (mAb) blockade or a colony stimulating factor 1 receptor inhibitor (CSF-1Ri) (PLX3397) and measured overall survival. We then harvested and characterized the PD-L1+ TIM population and its role in TIL activation and tumor cytolysis in vitro. Results: Our data indicated that the majority of PD-L1 expression in the GBM environment is contributed by TIMs rather than by tumor cells themselves. While PD-1 blockade partially reversed the TIL dysfunction, targeting TIMs directly with CSF-1Ri altered TIM expression of key chemotactic factors associated with promoting increased TIL infiltration after vaccination. Neither PD-1 mAb nor CSF-1Ri had a demonstrable therapeutic benefit alone, but when combined with DC vaccination, a significant survival benefit was observed. When the tripartite regimen was given (DC vaccine, PD-1 mAb, PLX3397), long-term survival was noted together with an increase in the number of TILs and TIL activation. Conclusion: Together, these studies elucidate the role that TIMs play in mediating adaptive immune resistance in the GBM microenvironment and provide evidence that they can be manipulated pharmacologically with agents that are clinically available. Development of immune resistance in response to active vaccination in GBM can be reversed with dual administration of CSF-1Ri and PD-1 mAb.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Vacunas contra el Cáncer/administración & dosificación , Glioblastoma/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Células Mieloides/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Femenino , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/prevención & control , Humanos , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Células Tumorales Cultivadas , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Immunol ; 197(1): 179-87, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27217585

RESUMEN

Cancer immunotherapies are increasingly effective in the clinic, especially immune checkpoint blockade delivered to patients who have T cell-infiltrated tumors. Agonistic CD40 mAb promotes stromal degradation and, in combination with chemotherapy, drives T cell infiltration and de novo responses against tumors, rendering resistant tumors susceptible to current immunotherapies. Partnering anti-CD40 with different treatments is an attractive approach for the next phase of cancer immunotherapies, with a number of clinical trials using anti-CD40 combinations ongoing, but the optimal therapeutic regimens with anti-CD40 are not well understood. Pancreatic ductal adenocarcinoma (PDA) is classically resistant to immunotherapy and lacks baseline T cell infiltration. In this study, we used a tumor cell line derived from a genetically engineered mouse model of PDA to investigate alterations in the sequence of anti-CD40 and chemotherapy as an approach to enhance pharmacological delivery of chemotherapy. Unexpectedly, despite our previous studies showing anti-CD40 treatment after chemotherapy is safe in both mice and patients with PDA, we report in this article that anti-CD40 administration <3 d in advance of chemotherapy is lethal in more than half of treated C57BL/6 mice. Anti-CD40 treatment 2 or 3 d before chemotherapy resulted in significantly increased populations of both activated myeloid cells and macrophages and lethal hepatotoxicity. Liver damage was fully abrogated when macrophage activation was blocked using anti-CSF-1R mAb. These studies highlight the dual nature of CD40 in activating both macrophages and T cell responses, and the need for preclinical investigation of optimal anti-CD40 treatment regimens for safe design of clinical trials.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD40/agonistas , Carcinoma Ductal/terapia , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Inmunoterapia/métodos , Fallo Hepático/prevención & control , Neoplasias Pancreáticas/terapia , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Animales , Carcinoma Ductal/complicaciones , Carcinoma Ductal/inmunología , Línea Celular Tumoral , Protocolos Clínicos , Interacciones Farmacológicas , Quimioterapia , Ingeniería Genética , Humanos , Fallo Hepático/etiología , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología
16.
Arthritis Res Ther ; 18: 75, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27036883

RESUMEN

BACKGROUND: CSF-1 or IL-34 stimulation of CSF1R promotes macrophage differentiation, activation and osteoclastogenesis, and pharmacological inhibition of CSF1R is beneficial in animal models of arthritis. The objective of this study was to determine the relative contributions of CSF-1 and IL-34 signaling to CSF1R in RA. METHODS: CSF-1 and IL-34 were detected by immunohistochemical and digital image analysis in synovial tissue from 15 biological-naïve rheumatoid arthritis (RA) , 15 psoriatic arthritis (PsA) and 7 osteoarthritis (OA) patients . Gene expression in CSF-1- and IL-34-differentiated human macrophages was assessed by FACS analysis and quantitative PCR. RA synovial explants were incubated with CSF-1, IL-34, control antibody (Ab), or neutralizing/blocking Abs targeting CSF-1, IL-34, or CSF1R. The effect of a CSF1R-blocking Ab was examined in murine collagen-induced arthritis (CIA). RESULTS: CSF-1 (also known as M-CSF) and IL-34 expression was similar in RA and PsA synovial tissue, but lower in controls (P < 0.05). CSF-1 expression was observed in the synovial sublining, and IL-34 in the sublining and the intimal lining layer. CSF-1 and IL-34 differentially regulated the expression of 17 of 336 inflammation-associated genes in macrophages, including chemokines, extra-cellular matrix components, and matrix metalloproteinases. Exogenous CSF-1 or IL-34, or their independent neutralization, had no effect on RA synovial explant IL-6 production. Anti-CSF1R Ab significantly reduced IL-6 and other inflammatory mediator production in RA synovial explants, and paw swelling and joint destruction in CIA. CONCLUSIONS: Simultaneous inhibition of CSF1R interactions with both CSF-1 and IL-34 suppresses inflammatory activation of RA synovial tissue and pathology in CIA, suggesting a novel therapeutic strategy for RA.


Asunto(s)
Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Animales , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Artritis Experimental/patología , Artritis Reumatoide/metabolismo , Benzo(a)Antracenos , Femenino , Citometría de Flujo , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucinas/biosíntesis , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones SCID , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo
17.
Eur J Haematol ; 96(4): 425-34, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26115424

RESUMEN

BACKGROUND: Multipotent mesenchymal stromal cells (MSCs) are used for prophylaxis of acute graft-versus-host disease (aGvHD) after allogeneic hematopoietic cell transplantation (allo-HCT). Not all samples of MSC are efficient for aGvHD prevention. The suitability of MSCs for aGvHD prophylaxis was studied. METHODS: MSCs were derived from the bone marrow (BM) of HCT donor and cultivated for no more than three passages. The characteristics of donor BM samples including colony-forming unit fibroblast (CFU-F) concentration, growth parameters of MSCs, and the relative expression levels (REL) of different genes were analyzed. MSCs were injected intravenously precisely at the moment of blood cell reconstitution. RESULTS: MSCs infusion induced a significant threefold decrease in aGvHD development and improved overall survival compared with the standard prophylaxis group. In ineffective MSC samples (9.4%), a significant decrease in total cell production and the REL of CSF1, FGFR1, and PDGFRB was observed. In all studied BM samples, the cumulative MSC production and CFU-F concentrations decreased with age. The expression levels of FGFR2, PPARG, and VEGF differed by age. CONCLUSIONS: A universal single indicator for the prediction of MSC eligibility for aGvHD prophylaxis was not identified. A multiparameter mathematical model for selecting MSC samples effective for the prevention of aGvHD was proposed.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Agonistas Mieloablativos/uso terapéutico , Acondicionamiento Pretrasplante/métodos , Adolescente , Adulto , Femenino , Expresión Génica , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/mortalidad , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Masculino , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/mortalidad , Síndromes Mielodisplásicos/patología , Síndromes Mielodisplásicos/terapia , PPAR gamma/genética , PPAR gamma/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/inmunología , Análisis de Supervivencia , Trasplante Homólogo
18.
J Clin Invest ; 125(10): 3952-64, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26389674

RESUMEN

Cytokines and metabolic pathway-controlling enzymes regulate immune responses and have potential as powerful tools to mediate immune tolerance. Blockade of the interaction between CD40 and CD40L induces long-term cardiac allograft survival in rats through a CD8+CD45RClo Treg potentiation. Here, we have shown that the cytokine IL-34, the immunoregulatory properties of which have not been previously studied in transplantation or T cell biology, is expressed by rodent CD8+CD45RClo Tregs and human FOXP3+CD45RCloCD8+ and CD4+ Tregs. IL-34 was involved in the suppressive function of both CD8+ and CD4+ Tregs and markedly inhibited alloreactive immune responses. Additionally, in a rat cardiac allograft model, IL-34 potently induced transplant tolerance that was associated with a total inhibition of alloantibody production. Treatment of rats with IL-34 promoted allograft tolerance that was mediated by induction of CD8+ and CD4+ Tregs. Moreover, these Tregs were capable of serial tolerance induction through modulation of macrophages that migrate early to the graft. Finally, we demonstrated that human macrophages cultured in the presence of IL-34 greatly expanded CD8+ and CD4+ FOXP3+ Tregs, with a superior suppressive potential of antidonor immune responses compared with non-IL-34-expanded Tregs. In conclusion, we reveal that IL-34 serves as a suppressive Treg-specific cytokine and as a tolerogenic cytokine that efficiently inhibits alloreactive immune responses and mediates transplant tolerance.


Asunto(s)
Interleucinas/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante , Traslado Adoptivo , Aloinjertos/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Dependovirus/genética , Factores de Transcripción Forkhead/análisis , Vectores Genéticos , Células HEK293 , Trasplante de Corazón , Humanos , Interleucinas/biosíntesis , Interleucinas/genética , Interleucinas/farmacología , Antígenos Comunes de Leucocito/análisis , Activación de Linfocitos , Factor Estimulante de Colonias de Macrófagos/fisiología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas Lew , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Proteínas Recombinantes de Fusión/genética , Análisis de Matrices Tisulares , Transducción Genética
19.
Lancet Oncol ; 16(8): 949-56, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26179200

RESUMEN

BACKGROUND: Diffuse-type tenosynovial giant cell tumour (dt-GCT) of the soft tissue (alternatively known as pigmented villonodular synovitis), an orphan disease with unmet medical need, is characterised by an overexpression of colony-stimulating factor 1 (CSF1), and is usually caused by a chromosomal translocation involving CSF1. CSF1 receptor (CSF1R) activation leads to the recruitment of CSF1R-expressing cells of the mononuclear phagocyte lineage that constitute the tumor mass in dt-GCT. Emactuzumab (RG7155) is a novel monoclonal antibody that inhibits CSF1R activation. We have assessed the safety, tolerability and activity of emactuzumab in patients with Dt-GCT of the soft tissue. METHODS: In this phase 1, first-in-human dose-escalation and dose-expansion study, eligible patients were aged 18 years or older with dt-GCT of the soft tissue with locally advanced disease or resectable tumours requiring extensive surgery, an Eastern Cooperative Oncology Group performance status of 1 or less, measurable disease according to Response Evaluation Criteria In Solid Tumors version 1.1, and adequate end-organ function. Patients with GCT of the bone were not eligible. Patients received intravenous emactuzumab at 900 mg, 1350 mg, or 2000 mg every 2 weeks in the dose-escalation phase and at the optimal biological dose in a dose-expansion phase. The primary objective was to evaluate the safety and tolerability of emactuzumab, and to determine the maximum tolerated dose or optimal biological dose. All treated patients were included in the analyses. Expansion cohorts are currently ongoing. This study is registered with ClinicalTrials.gov, number NCT01494688. FINDINGS: Between July 26, 2012, and Oct 21, 2013, 12 patients were enrolled in the dose-escalation phase. No dose-limiting toxicities were noted in the dose-escalation cohort; on the basis of pharmacokinetic, pharmacodynamic, and safety information, we chose a dose of 1000 mg every 2 week for the dose-expansion cohort, into which 17 patients were enrolled. Owing to different cutoff dates for safety and efficacy readouts, the safety population comprised 25 patients. Common adverse events after emactuzumab treatment were facial oedema (16 [64%] of 25 patients), asthenia (14 [56%]), and pruritus (14 [56%]). Five serious adverse events (periorbital oedema, lupus erythematosus [occurring twice], erythema, and dermohypodermitis all experienced by one [4%] patient each) were reported in five patients. Three of the five serious adverse events-periorbital oedema (one [4%]), lupus erythematosus (one [4%]), and dermohypodermitis (one [4%])-were assessed as grade 3. Two other grade 3 events were reported: mucositis (one [4%]) and fatigue (one [4%]). 24 (86%) of 28 patients achieved an objective response; two (7%) patients achieved a complete response. INTERPRETATION: Further study of dt-GCT is warranted and different possibilities, such as an international collaboration with cooperative groups to assure appropriate recruitment in this rare disease, are currently being assessed. FUNDING: F Hoffmann-La Roche.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antineoplásicos/administración & dosificación , Tumores de Células Gigantes/tratamiento farmacológico , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Sinovitis Pigmentada Vellonodular/tratamiento farmacológico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Antineoplásicos/efectos adversos , Esquema de Medicación , Femenino , Tumores de Células Gigantes/inmunología , Tumores de Células Gigantes/metabolismo , Tumores de Células Gigantes/patología , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias de los Tejidos Blandos/inmunología , Neoplasias de los Tejidos Blandos/metabolismo , Neoplasias de los Tejidos Blandos/patología , Sinovitis Pigmentada Vellonodular/inmunología , Sinovitis Pigmentada Vellonodular/metabolismo , Sinovitis Pigmentada Vellonodular/patología , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
20.
J Leukoc Biol ; 98(4): 641-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26136505

RESUMEN

Although Mϕ represent the most primordial immune cell subsets, the mechanisms governing their functional heterogeneity remain poorly defined. However, it is well established that the CSF-1 cytokine contributes to monopoiesis and to this heterogeneity, whereas the unrelated IL-34 also binds the CSF-1R toward poorly understood immunologic roles. To delineate the molecular and evolutionary basis behind vertebrate Mϕ functional heterogeneity, we performed comprehensive transcriptional and functional studies of amphibian (Xenopus laevis) BM (in vitro) and PER (in vivo) Mϕ derived by rXlCSF-1 and rXlIL-34. Our findings indicate that these amphibian cytokines promote morphologically and functionally distinct Mϕ populations. Mϕ induced by rXlCSF-1 possess more robust iNOS gene expression, are substantially more phagocytic, display greater NO responses, and exhibit enhanced bactericidal capacities. By contrast, rXlIL-34-derived Mϕ express greater levels of Arg-1 and NADPH oxidase components and possess greater respiratory burst responses. Most notably, whereas CSF-1 Mϕ are highly susceptible to the emerging FV3 ranavirus, rXlIL-34 Mϕ exhibit potent antiviral activity against this pathogen, which is dependent on reactive oxygen production. This work marks an advance in our understanding of the possible mechanisms governing vertebrate Mϕ functional heterogeneity.


Asunto(s)
Interleucinas/inmunología , Macrófagos/citología , Macrófagos/inmunología , Animales , Células Cultivadas , Factor Estimulante de Colonias de Macrófagos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...