Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
1.
J Clin Invest ; 133(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37490346

RESUMEN

Sickle cell disease (SCD) is a hereditary hemoglobinopathy characterized by painful vaso-occlusive crises (VOC) and chronic hemolysis. The mononuclear phagocyte system is pivotal to SCD pathophysiology, but the mechanisms governing monocyte/macrophage differentiation remain unknown. This study examined the influence of hemolysis on circulating monocyte trajectories in SCD. We discovered that hemolysis stimulated CSF-1 production, partly by endothelial cells via Nrf2, promoting classical monocyte (CMo) differentiation into blood patrolling monocytes (PMo) in SCD mice. However, hemolysis also upregulated CCL-2 through IFN-I, inducing CMo transmigration and differentiation into tissue monocyte-derived macrophages. Blocking CMo transmigration by anti-P selectin antibody in SCD mice increased circulating PMo, corroborating that CMo-to-tissue macrophage differentiation occurs at the expense of CMo-to-blood PMo differentiation. We observed a positive correlation between plasma CSF-1/CCL-2 ratios and blood PMo levels in patients with SCD, underscoring the clinical significance of these two opposing factors in monocyte differentiation. Combined treatment with CSF-1 and anti-P selectin antibody more effectively increased PMo numbers and reduced stasis compared with single-agent therapies in SCD mice. Altogether, these data indicate that monocyte fates are regulated by the balance between two heme pathways, Nrf2/CSF-1 and IFN-I/CCL-2, and suggest that the CSF-1/CCL-2 ratio may present a diagnostic and therapeutic target in SCD.


Asunto(s)
Anemia de Células Falciformes , Enfermedades Vasculares , Ratones , Animales , Hemólisis , Monocitos/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Células Endoteliales/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/tratamiento farmacológico , Enfermedades Vasculares/metabolismo , Diferenciación Celular , Selectinas/metabolismo , Selectinas/uso terapéutico
2.
BMC Pulm Med ; 23(1): 53, 2023 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-36737697

RESUMEN

BACKGROUND: Patients with tuberculosis (TB) and HIV often present with impairments in lung function and exercise capacity after treatment. We evaluated clinical and immunologic variables associated with a minimum clinically important difference (MCID) in the change in the 6 min walk test distance during the first 24 weeks of antiretroviral (ART) and anti-tubercular therapy. METHODS: Adults initiating ART and anti-TB treatment in the setting of newly-diagnosed HIV and pulmonary TB were enrolled in a prospective cohort study in South Africa. Patients underwent 6 min walk tests and spirometry at weeks 0, 4, 12, and 24 and biomarker level measurements early during treatment, at weeks 0, 4, and 12, when inflammation levels are typically elevated. Biomarkers included matrix metalloproteinases-1 (MMP-1), tissue inhibitor of MMP (TIMP)-1, collagen 1a, IL-6, IL-8, vascular cell adhesion molecule 1 (VCAM-1), C-X-C motif chemokine 10 (CXCL-10), CXCL-11, macrophage colony-stimulating factor (M-CSF), plasminogen activator, vascular endothelial growth factor, and chemokine (C-C) motif-2 (CCL-2). An MCID was derived statistically, and achievement of an MCID was modeled as the outcome using logistic regression model. RESULTS: Eighty-nine patients walked an average of 393 (± standard deviation = 69) meters at baseline, which increased by an average of 9% (430 ± 70 m) at week 24. The MCID for change in walk distance was estimated as 41 m. Patients experiencing an MCID on treatment had worse lung function, lower 6 min walk test distance, higher levels of proinflammatory biomarkers including TIMP-1 and M-CSF, and lower levels of collagen 1a at baseline. Experiencing an MCID during treatment was associated with increases in forced expiratory volume in 1-s [odds ratio (OR) = 1.17, 95% confidence interval (CI) = 1.05-1.33] and increases in blood collagen 1a levels (OR = 1.31, 95%CI 1.06-1.62). CONCLUSIONS: ART and TB treatment are associated with substantial improvements in 6 min walk test distance over time. Achievement of an MCID in the 6 min walk test in this study was associated with more severe disease at baseline and increases in collagen 1a levels and lung function during therapy.


Asunto(s)
Infecciones por VIH , Tuberculosis , Humanos , Adulto , Prueba de Paso , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Estudios Prospectivos , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Tuberculosis/complicaciones , Biomarcadores , Pulmón , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico
3.
Technol Health Care ; 31(2): 635-645, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36314174

RESUMEN

BACKGROUND: Glioblastoma is the most common and most aggressive type of primary brain tumor. OBJECTIVE: The aim of this study was to investigate the efficacy and safety of intranasal granulocyte-macrophage colony stimulating factor (GM-CSF) administration combined with chemoradiotherapy in patients with glioblastoma who underwent surgery. METHODS: Ninety-two patients were randomly divided into two groups: a control group (n= 46), who received radiotherapy with adjuvant local delivery of nimustine hydrochloride (ACNU) and systemic administration of temozolomide, and an intervention group (n= 46), who received intranasal GM-CSF prior to each cycle of adjuvant chemotherapy in addition to the treatment of the control group. Karnofsky performance status (KPS) scores, progression-free survival (PFS), overall survival (OS), and adverse effects were calculated and compared between the two groups. RESULTS: Compared with the control group, the intervention group had longer PFS (7.8 vs. 6.9 months, P= 0.016) and OS (19.2 vs. 17.1 months, P= 0.045, without adjustment for interim analyses). The KPS scores were also higher in the intervention group than in the control group after 6 months (84.35 ± 8.86 vs. 80.65 ± 7.72; t= 4.552, P= 0.036). Furthermore, the patients in the intervention group had lower incidence of neutropenia and thrombocytopenia (8.7% vs. 29.5%, P= 0.012; 8.7% vs. 18.2%, P= 0.186). Other adverse events were similar in both groups, and most adverse events were grade I/II and resolved spontaneously. CONCLUSION: Intranasal GM-CSF enhances the efficacy of the local ACNU administration combined with oral temozolomide chemotherapy. The survival and performance status were significantly improved in patients with glioblastoma after surgery. Additionally, the GM-CSF therapy was able to reduce the occurrence of chemotherapy-related neutropenia and thrombocytopenia.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neutropenia , Trombocitopenia , Humanos , Temozolomida/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Nimustina/uso terapéutico , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Neutropenia/tratamiento farmacológico , Trombocitopenia/tratamiento farmacológico , Granulocitos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia
4.
Folia Med Cracov ; 62(2): 93-107, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36256897

RESUMEN

Tenosynovial Giant Cell Tumor (TGCT) is a group of typically benign lesions arising from the synovium of joints, bursae and tendon sheaths. Depending on their growth pattern and clinical course, they are divided into localized and diffuse types. It is predominantly caused by a mutation in the stromal cells of the synovial membrane leading to overexpression of the colony stimulating factor 1 that recruits CSF1R-expressing cells of the mononuclear phagocyte lineage into the tumor mass. The lesions contain mainly histiocyte-like and synovial cells accompanied by varying numbers of multinucleated giant cells, mononuclear cells, foam cells, inflammatory cells and hemosiderin deposits. The gold standard for detect- ing and monitoring the disease is MRI, where the characteristic hemosiderin accumulation can be best appreciated, but it is a histological examination that is most conclusive. The main treatment is surgical resection of all pathological tissue, but radio- and chemotherapy are also viable options for certain groups of patients.


Asunto(s)
Tumor de Células Gigantes de las Vainas Tendinosas , Tumores de Células Gigantes , Sinovitis Pigmentada Vellonodular , Humanos , Sinovitis Pigmentada Vellonodular/terapia , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Tumores de Células Gigantes/tratamiento farmacológico , Tumores de Células Gigantes/patología , Tumores de Células Gigantes/cirugía , Hemosiderina/uso terapéutico
5.
Pharmacol Res ; 184: 106400, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35988868

RESUMEN

BACKGROUND AND OBJECTIVE: Bone loss occurs in several inflammatory diseases because of chronic persistent inflammation that activates osteoclasts (OCs) to increase bone resorption. Currently available antiresorptive drugs have severe side effects or contraindications. Herein, we explored the effects and mechanism of Alpinetin (Alp) on receptor activator of nuclear factor κB ligand (RANKL)-mediated OCs differentiation, function, and in inflammatory osteolysis of mice. METHOD: Primary mouse bone marrow-derived macrophages (BMMs) induced by RANKL and macrophage colony-stimulating factor (M-CSF) were utilized to test the impact of Alp on OCs differentiation, function, and intracellular reactive oxygen species (ROS) production, respectively. Expression of oxidant stress relevant factors and OCs specific genes were assessed via real-time quantitative PCR. Further, oxidative stress-related factors, NF-κB, MAPK, PI3K/AKT/GSK3-ß, and NFATc1 pathways were examined via Western blot. Finally, LPS-induced mouse calvarial osteolysis was used to investigate the effect of Alp on inflammatory osteolysis in vivo. RESULT: Alp suppressed OCs differentiation and resorption function, and down-regulated the ROS production. Alp inhibited IL-1ß, TNF-α and osteoclast-specific gene transcription. It also blocked the gene and protein expression of Nox1 and Keap1, but enhanced Nrf2, CAT, and HO-1 protein levels. Additionally, Alp suppressed the phosphorylation of PI3K and P38, and restrained the expression of osteoclast-specific gene Nfatc1 and its auto-amplification, hence minimizing LPS-induced osteolysis in mice. CONCLUSION: Alp is a novel candidate or therapeutics for the osteoclast-associated inflammatory osteolytic ailment.


Asunto(s)
Conservadores de la Densidad Ósea , Osteólisis , Animales , Conservadores de la Densidad Ósea/farmacología , Diferenciación Celular , Flavanonas , Glucógeno Sintasa Quinasa 3/metabolismo , Inflamación/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Lipopolisacáridos/farmacología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Osteoclastos , Osteólisis/inducido químicamente , Osteólisis/tratamiento farmacológico , Oxidantes/metabolismo , Oxidantes/farmacología , Oxidantes/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ligando RANK/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
6.
Br J Cancer ; 127(6): 1142-1152, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35725813

RESUMEN

BACKGROUND: Disseminated BRAFV600E melanoma responds to BRAF inhibitors (BRAFi) but easily develops resistance with poor prognosis. Secretome plays a pivotal role during tumour progression causing profound effects on therapeutic efficacy. Secreted M-CSF is involved in both cytotoxicity suppression and tumour progression in melanoma. We aimed to analyse the M-CSF contribution in resistant metastatic melanoma to BRAF-targeted therapies. METHODS: Conditioned media from melanoma cells were analysed by citoarray. Viability and migration/invasion assays were performed with paired melanoma cells and tumour growth in xenografted SCID mice. We evaluated the impact of M-CSF plasma levels with clinical prognosis from 35 metastatic BRAFV600E-mutant melanoma patients. RESULTS: BRAFi-resistant melanoma cells secretome is rich in pro-tumour cytokines. M-CSF secretion is essential to induce a Vemurafenib-resistant phenotype in melanoma cells. Further, we demonstrated that M-CSF mAb in combination with Vemurafenib and autophagy blockers synergistically induce apoptosis, impair migration and reduce tumour growth in BRAFi-resistant melanoma cells. Interestingly, lower M-CSF plasma levels are associated with better prognosis in metastatic melanoma patients. CONCLUSIONS: Secreted M-CSF induces a BRAFi-resistant phenotype and means worse prognosis in BRAFV600E metastatic melanoma patients. These results identify secreted M-CSF as a promising therapeutic target toward BRAFi-resistant melanomas.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas B-raf , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Indoles/farmacología , Indoles/uso terapéutico , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/farmacología , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Ratones , Ratones SCID , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Sulfonamidas/farmacología , Vemurafenib/farmacología , Vemurafenib/uso terapéutico
7.
Neurourol Urodyn ; 41(3): 787-796, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35170790

RESUMEN

INTRODUCTION: Bladder outlet obstruction (BOO) is a common problem that can affect bladder structure and function. Currently, there is no effective drugs available to prevent BOO-induced remodeling. Previous reports have demonstrated that the pathogenesis of BOO is associated with macrophage infiltration and polarization, which is physiologically dependent on colony-stimulating factor 1 receptor (CSF-1R) activation. Here we utilized a highly selective CSF-1R inhibitor, GW2580, to determine its preventive effects on BOO-induced remodeling. METHODS: A total of 24 Sprague-Dawley rats were randomly divided into sham, BOO + vehicle, and BOO + GW2580 group. GW2580 or vehicle control was administrated by oral gavage at daily doses of 40 mg/kg for 6 weeks. Bladder samples were collected for histopathology, immunohistochemistry, immunofluorescence, western blotting, and flow cytometry analysis. RESULTS: Our results demonstrated that bladder fibrosis was ameliorated by GW2580 compared with the vehicle group (22.01% ± 5.13% vs. 32.15% ± 7.24%, p < 0.01). Furthermore, treatment with GW2580 induced an inhibition of macrophage infiltration (4.41% ± 1.28% vs. 13.57% ± 3.42%, p < 0.001) and M2 macrophage polarization (10.67% ± 4.15% vs. 28.59% ± 6.38%, p < 0.001). There was also a decrease of profibrotic F4/80+  α-smooth muscle actin+ (α-SMA+ ) macrophage to myofibroblast transition (9.11% ± 2.58% vs. 17.33% ± 4.01%, p < 0.001) and CD163+ TGF-ß1+ cells (7.68% ± 2.10% vs. 14.17% ± 4.09%, p < 0.01) in the GW2580 group when compared with the vehicle group. CONCLUSIONS: In summary, our findings showed that GW2580 is a worthwhile candidate for a follow-up study to test in the treatment of BOO-induced remodeling.


Asunto(s)
Obstrucción del Cuello de la Vejiga Urinaria , Animales , Modelos Animales de Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Masculino , Ratas , Ratas Sprague-Dawley , Vejiga Urinaria
8.
Clin Infect Dis ; 75(2): 278-287, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34718451

RESUMEN

BACKGROUND: Neutralizing anti-granulocyte-macrophage colony-stimulating factor (GM-CSF) autoantibodies (AAbs) have been increasingly recognized to predispose healthy individuals to disseminated cryptococcosis. However, studies have only considered patients with central nervous system (CNS) infection. No longitudinal study has captured the disease spectrum and clinical course. METHODS: We prospectively enrolled adults without human immunodeficiency virus infection who had disseminated or unusual cryptococcosis. We compared the demographics, clinical features, kinetics of serum cryptococcal antigen (CrAg) titers, anti-GM-CSF AAb concentrations, and treatment outcomes between patients with (case patients) and without (control patients) anti-GM-CSF AAbs. Additional reports from the literature were also reviewed. RESULTS: Twenty-three patients were enrolled, of whom 6 tested positive for anti-GM-CSF AAbs. All case patients with positive fungal cultures (5/5 [100%]) were infected with Cryptococcus gattii VGII. Among them, 3 had exclusively pulmonary involvement, and 1 had only musculoskeletal lesions. Patients with CNS cryptococcosis exhibited a higher serum concentration of anti-GM-CSF AAbs than those with extraneural cryptococcosis. Case patients had higher initial and peak levels of serum CrAg and longer duration of antigenemia compared with the control patients. All case patients who had completed antifungal therapy had favorable outcomes without recurrence. CONCLUSIONS: Testing for anti-GM-CSF AAbs should be considered for not only previously healthy patients with disseminated cryptococcosis but also those with unexplained, localized cryptococcosis. Recurrence after completion of antifungal therapy was rare despite the persistence of anti-GM-CSF AAbs.


Asunto(s)
Autoanticuerpos , Criptococosis , Adulto , Antifúngicos/uso terapéutico , Antígenos Fúngicos , Sistema Nervioso Central , Criptococosis/diagnóstico , Criptococosis/tratamiento farmacológico , Estudios de Seguimiento , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Factor Estimulante de Colonias de Macrófagos/uso terapéutico
9.
J Pediatr Hematol Oncol ; 43(3): e358-e364, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31815885

RESUMEN

Japanese patients with neuroblastoma completing induction therapy and high-dose chemotherapy received antidisialoganglioside antibody dinutuximab 17.5 mg/m2 for 4 days during each of 5 consecutive 28-day cycles. Patients also received macrophage colony-stimulating factor (M-CSF) or granulocyte colony-stimulating factor (G-CSF) during cycles 1, 3, and 5 combined with interleukin-2 teceleukin during cycles 2 and 4. A total of 25 patients (11 in the M-CSF group and 14 in the G-CSF group) were enrolled, and dose-limiting toxicity was assessed in the first 12 patients (6 in each group). The recommended doses of dinutuximab, M-CSF, and G-CSF were determined to be 17.5 mg/m2, 6.0×106 U/m2, and 5 µg/kg/d, respectively, whereas that of teceleukin was 0.75×106 IU/m2 during week 1 and 1×106 IU/m2 during week 2. The most common grade 3 or 4 adverse events in both groups were neutrophil count decreased, platelet count decreased, pyrexia, and alanine aminotransferase increased. Four patients (2 in each group) discontinued the treatment because of adverse events. At the end of the study, survival was confirmed in 22 patients (9 in the M-CSF group and 13 in the G-CSF group). From these results, we concluded that this combination regimen is a feasible treatment for Japanese patients with neuroblastoma.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Adolescente , Anticuerpos Monoclonales/efectos adversos , Antineoplásicos Inmunológicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Preescolar , Femenino , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Interleucina-2/efectos adversos , Interleucina-2/uso terapéutico , Japón/epidemiología , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Masculino , Neuroblastoma/epidemiología , Resultado del Tratamiento
10.
Clin Infect Dis ; 73(7): e2465-e2469, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32894278

RESUMEN

BACKGROUND: The treatment of cutaneous leishmaniasis (CL) in Brazil using pentavalent antimony (Sbv) is associated with a high rate of failure. Miltefosine has proven efficacy for CL caused by L. braziliensis, with a cure rate (CR) of 75%. A combined treatment with granulocyte macrophage colony-stimulating factor (GM-CSF) and miltefosine could increase CR and decrease healing time. METHODS: A randomized, double-blind clinical trial to evaluate the efficacy of miltefosine combined with topical GM-CSF (M + GM) vs miltefosine and placebo (M + P) vs Sbv in 133 patients with CL caused by L. braziliensis in Bahia, Brazil. RESULTS: The final CR at 180 days after the initiation of treatment was 44.4% in the Sbv group, 76.6% in the M + P group (P = .003 vs Sbv), and 75.6% in the M + GM group (P = .004 vs Sbv). The median healing time for cure was 102 days for the Sbv group and 60 days for both miltefosine groups (P = .0009). During the 6-month follow-up period, 4 relapses were documented: 1 in the Sbv group, 1 in the M + P group, and 2 in the M + GM group. Mild adverse events occurred in 65% of patients from the Sbv group, 76% and 79% from the M + P and M + GM groups respectively. CONCLUSIONS: Miltefosine is more effective than Sbv for the treatment of CL caused by L. braziliensis in Brazil and accelerates the healing time. Association with GM-CSF does not improve therapeutic outcome. CLINICAL TRIALS REGISTRATION: NCT03023111.


Asunto(s)
Antiprotozoarios , Leishmania braziliensis , Leishmaniasis Cutánea , Antimonio/uso terapéutico , Antiprotozoarios/uso terapéutico , Brasil , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Granulocitos , Humanos , Leishmaniasis Cutánea/tratamiento farmacológico , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Fosforilcolina/análogos & derivados , Resultado del Tratamiento
11.
Int J Infect Dis ; 103: 358-363, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33253864

RESUMEN

OBJECTIVES: To compare topical granulocyte and macrophage colony-stimulating factor (GM-CSF) and miltefosine (G + M) versus placebo and miltefosine (P + M) or parenteral meglumine antimoniate (MA) in the treatment of 150 patients with cutaneous leishmaniasis (CL) caused by Leishmania guyanensis in the Amazon. DESIGN: A randomized and double-blinded clinical trial. RESULTS: At 90 days after the initiation of therapy, the cure rates were 66%, 58%, and 52% for the groups P + M, G + M, and MA, respectively (p > 0.05). Cure rates at 180 days did not differ. Healing time was similar in the 3 groups, but faster in the MA group as compared to the G + M group (p = 0.04). Mild and transitory systemic adverse events were frequent in all groups (above 85%). Nausea (85%) and vomiting (39%) predominated in the miltefosine groups and arthralgia (51%) and myalgia (48%) in the MA group. One patient (group MA) stopped treatment after presenting with fever, exanthema, and severe arthralgia. CONCLUSIONS: Miltefosine did not present a higher cure rate than MA, and the association of GM-CSF did not improve the therapeutic response. Nevertheless, because of its less toxicity, easier administration, and a similar cure rate when compared with MA, miltefosine should remain as one of the main drugs for treating CL due to L. guyanensis. (Clinicaltrials.gov Identifier NCT03023111).


Asunto(s)
Antiprotozoarios/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Leishmaniasis Cutánea/tratamiento farmacológico , Antimoniato de Meglumina/uso terapéutico , Fosforilcolina/análogos & derivados , Administración Oral , Administración Tópica , Adolescente , Adulto , Antiprotozoarios/administración & dosificación , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Humanos , Factor Estimulante de Colonias de Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Masculino , Antimoniato de Meglumina/administración & dosificación , Persona de Mediana Edad , Fosforilcolina/administración & dosificación , Fosforilcolina/uso terapéutico , Adulto Joven
12.
Cell Death Dis ; 10(5): 353, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31040267

RESUMEN

A variety of osteolytic factors have been identified from breast cancer cells leading to osteolysis, but less is known about which factor plays an essential role in the initiation process prior to the overt vicious osteolytic cycle. Here, we present in vitro and in vivo evidences to clarify the role of interleukin-11 (IL-11) as an essential contributor to breast cancer bone metastasis mediated osteolysis. Animal studies showed that bone specific metastatic BoM-1833 cells induce earlier onset of osteolysis and faster tumor growth compared with MCF7 and parental MDA-MB-231 cells in BALB/c-nu/nu nude mice. IL-11 was further screened and identified as the indispensable factor secreted by BoM-1833 cells inducing osteoclastogenesis independently of receptor activator of nuclear factor κB ligand (RANKL). Mechanistic investigation revealed that the JAK1/STAT3 signaling pathway as a downstream effector of IL-11, STAT3 activation further induces the expression of c-Myc, a necessary factor required for osteoclastogenesis. By inhibiting STAT3 phosphorylation, AG-490 was shown effective in reducing osteolysis and tumor growth in the metastatic niche. Overall, our results revealed the essential role and the underlying molecular mechanism of IL-11 in breast cancer bone metastasis mediated osteolysis. STAT3 targeting through AG-490 is a potential therapeutic strategy for mitigating osteolysis and tumor growth of bone metastatic breast cancer.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias de la Mama/patología , Interleucina-11/farmacología , Osteogénesis/efectos de los fármacos , Osteólisis/prevención & control , Ligando RANK/farmacología , Animales , Neoplasias Óseas/mortalidad , Neoplasias Óseas/secundario , Huesos/diagnóstico por imagen , Huesos/patología , Línea Celular Tumoral , Femenino , Humanos , Interleucina-11/metabolismo , Interleucina-11/uso terapéutico , Janus Quinasa 1/metabolismo , Estimación de Kaplan-Meier , Factor Estimulante de Colonias de Macrófagos/farmacología , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Osteólisis/patología , Ligando RANK/uso terapéutico , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología , Tirfostinos/uso terapéutico
13.
Thromb Haemost ; 119(2): 234-245, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30650445

RESUMEN

Macrophages make important contributions to inflammation and wound healing. We show here that macrophage polarization is deregulated in haemophilia in response to macrophage colony-stimulating factor (M-CSF) and partially in response to granulocyte-macrophage colony-stimulating factor (GM-CSF). As a result, haemophilia macrophages exhibit a specific impairment of M-CSF-mediated functions involved in wound healing such as clot invasion and phagocytosis. Haemophilia monocytes express reduced amounts of the receptors for M-CSF and GM-CSF, which correlates with a failure to express tumour necrosis factor α (TNFα) and CD163 in M-CSF-treated haemophilia macrophages and reduced expression of TNFα and CD206 after treatment with GM-CSF. Protein expression in response to M-CSF was regained with respect to CD163 and CD206 after embedding haemophilia monocytes in clotted plasma suggesting that a functioning coagulation system has positive effects on macrophage M2 polarization. Mimicking the functional deficits of haemophilia macrophages in normal macrophages was possible by adding leptin, which we found to be elevated in the blood of haemophilia patients, to a monocyte cell line. The increase of leptin occurred in conjunction with C-reactive protein in a body mass index-controlled cohort suggesting that haemophilia patients harbour chronic low-grade inflammation. Together, our data indicate that impaired clotting in haemophilia patients leads to increased inflammation and a deregulation in macrophage differentiation, which may explain the commonly observed deficits in wound healing and tissue regeneration.


Asunto(s)
Coagulación Sanguínea , Hemofilia A/sangre , Macrófagos/citología , Adulto , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Circulación Sanguínea , Diferenciación Celular , Estudios de Cohortes , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/citología , Fibrina/metabolismo , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Hemofilia A/tratamiento farmacológico , Humanos , Lectinas Tipo C/metabolismo , Leucocitos Mononucleares/citología , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Microscopía Fluorescente , Monocitos/citología , Fagocitosis , Receptores de Superficie Celular/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
14.
J Exp Med ; 213(11): 2269-2279, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27811055

RESUMEN

Myeloablative treatment preceding hematopoietic stem cell (HSC) and progenitor cell (HS/PC) transplantation results in severe myeloid cytopenia and susceptibility to infections in the lag period before hematopoietic recovery. We have previously shown that macrophage colony-stimulating factor (CSF-1; M-CSF) directly instructed myeloid commitment in HSCs. In this study, we tested whether this effect had therapeutic benefit in improving protection against pathogens after HS/PC transplantation. M-CSF treatment resulted in an increased production of mature myeloid donor cells and an increased survival of recipient mice infected with lethal doses of clinically relevant opportunistic pathogens, namely the bacteria Pseudomonas aeruginosa and the fungus Aspergillus fumigatus M-CSF treatment during engraftment or after infection efficiently protected from these pathogens as early as 3 days after transplantation and was effective as a single dose. It was more efficient than granulocyte CSF (G-CSF), a common treatment of severe neutropenia, which showed no protective effect under the tested conditions. M-CSF treatment showed no adverse effect on long-term lineage contribution or stem cell activity and, unlike G-CSF, did not impede recovery of HS/PCs, thrombocyte numbers, or glucose metabolism. These results encourage potential clinical applications of M-CSF to prevent severe infections after HS/PC transplantation.


Asunto(s)
Aspergilosis/tratamiento farmacológico , Aspergilosis/prevención & control , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/prevención & control , Animales , Aspergilosis/sangre , Aspergilosis/microbiología , Aspergillus/efectos de los fármacos , Aspergillus/fisiología , Glucemia/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones Endogámicos C57BL , Mielopoyesis/efectos de los fármacos , Infecciones por Pseudomonas/sangre , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología
15.
Cochrane Database Syst Rev ; 2: CD009310, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26880256

RESUMEN

BACKGROUND: Myelodysplastic syndromes (MDS) are a heterogeneous group of haematological diseases which are characterised by a uni- or multilineage dysplasia of haematological stem cells. Standard treatment is supportive care of the arising symptoms including red blood cell transfusions or the administration of erythropoiesis-stimulating agents (ESAs) in the case of anaemia or the treatment with granulocyte (G-CSF) and granulocyte-macrophage colony stimulating factors (GM-CSF) in cases of neutropenia. OBJECTIVES: The objective of this review is to assess the evidence for the treatment of patients with MDS with G-CSF and GM-CSF in addition to standard therapy in comparison to the same standard therapy or the same standard therapy and placebo. SEARCH METHODS: We searched MEDLINE (from 1950 to 3 December 2015) and CENTRAL (Cochrane Central Register of Controlled Trials until 3 December 2015), as well as conference proceedings (American Society of Hematology, American Society of Clinical Oncology, European Hematology Association, European Society of Medical Oncology) for randomised controlled trials (RCTs). Two review authors independently screened search results. SELECTION CRITERIA: We included RCTs examining G-CSF or GM-CSF in addition to standard therapy in patients with newly diagnosed MDS. DATA COLLECTION AND ANALYSIS: We used hazard ratios (HR) as effect measure for overall survival (OS), progression-free survival (PFS) and time to progression, and risk ratios for response rates, adverse events, antibiotic use and hospitalisation. Two independent review authors extracted data and assessed risk of bias. Investigators of two trials were contacted for subgroup information, however, no further data were provided. G-CSF and GM-CSF were analysed separately. MAIN RESULTS: We screened a total of 566 records. Seven RCTs involving 486 patients were identified, but we could only meta-analyse the two evaluating GM-CSF. We judged the potential risk of bias of these trials as unclear, mostly due to missing information. All trials were randomised and open-label studies. However, three trials were published as abstracts only, therefore we were not able to assess the potential risk of bias for these trials in detail. Overall, data were not reported in a comparable way and patient-related outcomes like survival, time to progression to acute myeloid leukaemia (AML) or the incidence of infections was reported in two trials only.Five RCTs (N = 337) assessed the efficacy of G-CSF in combination with standard therapy (supportive care, chemotherapy or erythropoietin). We were not able to perform meta-analyses for any of the pre-planned outcomes due to inconsistent and insufficient reporting of data. There is no evidence for a difference for overall survival (hazard ratio (HR) 0.80, 95% confidence interval (CI) 0.44 to 1.47), progression-free survival (only P value provided), progression to AML, incidence of infections and number of red blood transfusions (average number of 12 red blood cell transfusions in each arm). We judged the quality of evidence for all these outcomes as very low, due to very high imprecision and potential publication bias, as three trials were published as abstracts only. Data about quality of life and serious adverse events were not reported in any of the included trials.Two RCTs (N = 149) evaluated GM-CSF in addition to standard therapy (chemotherapy). For mortality (two RCTs; HR 0.88, 95% CI 0.62 to 1.26), we found no evidence for a difference (low-quality evidence). Data for progression-free survival and serious adverse events were not comparable across both studies, without evidence for a difference between both arms (low-quality evidence). For infections, red blood cell and platelet transfusions, we found no evidence for a difference, however, these outcomes were reported by one trial only (low-quality evidence). Time to progression to AML and quality of life were not reported at all.Moreover, we identified two cross-over trials, including 244 patients and evaluating GM-CSF versus placebo, without publishing results for each arm before crossing over. In addition, we identified two ongoing studies, one of which was discontinued due to withdrawal of pharmaceutical support, the other was terminated early, both without publishing results. AUTHORS' CONCLUSIONS: Although we identified seven trials with a total number of 486 patients, and two unpublished, prematurely finished studies, this systematic review mainly shows that there is a substantial lack of data, which might inform the use of G-CSF and GM-CSF for the prevention of infections, prolonging of survival and improvement of quality of life. The impact on progression to AML remains unclear.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Síndromes Mielodisplásicos/tratamiento farmacológico , Eritropoyetina/uso terapéutico , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto
16.
J Neurosci ; 35(3): 1136-48, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25609628

RESUMEN

Approaches to stimulate remyelination may lead to recovery from demyelinating injuries and protect axons. One such strategy is the activation of immune cells with clinically used medications, since a properly directed inflammatory response can have healing properties through mechanisms such as the provision of growth factors and the removal of cellular debris. We previously reported that the antifungal medication amphotericin B is an activator of circulating monocytes, and their tissue-infiltrated counterparts and macrophages, and of microglia within the CNS. Here, we describe that amphotericin B activates these cells through engaging MyD88/TRIF signaling. When mice were subjected to lysolecithin-induced demyelination of the spinal cord, systemic injections of nontoxic doses of amphotericin B and another activator, macrophage colony-stimulating factor (MCSF), further elevated the representation of microglia/macrophages at the site of injury. Treatment with amphotericin B, particularly in combination with MCSF, increased the number of oligodendrocyte precursor cells and promoted remyelination within lesions; these pro-regenerative effects were mitigated in mice treated with clodronate liposomes to reduce circulating monocytes and tissue-infiltrated macrophages. Our results have identified candidates among currently used medications as potential therapies for the repair of myelin.


Asunto(s)
Anfotericina B/farmacología , Enfermedades Desmielinizantes/tratamiento farmacológico , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Microglía/efectos de los fármacos , Monocitos/efectos de los fármacos , Vaina de Mielina/efectos de los fármacos , Regeneración Nerviosa/efectos de los fármacos , Anfotericina B/uso terapéutico , Animales , Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Enfermedades Desmielinizantes/inducido químicamente , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Macrófagos/fisiología , Ratones , Microglía/fisiología , Monocitos/fisiología , Vaina de Mielina/fisiología , Regeneración Nerviosa/fisiología
17.
Rev. patol. respir ; 17(2): 71-75, abr.-jun. 2014. ilus
Artículo en Español | IBECS | ID: ibc-123815

RESUMEN

Presentamos un caso de proteinosis alveolar pulmonar primaria en un varón de 46 años en el que la aproximación diagnóstica por criterios clínicos, técnicas de imagen, estudios complementarios y muestras obtenidas por broncofibroscopia (bioquímica, celularidad e histología) confirman el diagnóstico. Se plantean controversias con las opciones terapéuticas actuales relacionadas con el lavado terapéutico y el uso de factores estimulantes del crecimiento de macrófagos


We report a case of primary alveolar proteinosis in a 46 years man with a diagnostic approach based on clinical criteria, imaging, complementary studies and samples obtained by bronchoscopy (biochemistry, cellularity and histology) confirmed the diagnosis. Disputes arise with current therapeutic options related to therapeutic lavage and use of growth factors stimulating macrophages


Asunto(s)
Humanos , Masculino , Persona de Mediana Edad , Proteinosis Alveolar Pulmonar/terapia , Lavado Broncoalveolar , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Radiografía Torácica
18.
Pak J Pharm Sci ; 27(2): 245-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24577909

RESUMEN

Low doses of granulocyte- colony stimulating factor (G-CSF) and granulocyte macrophage- colony stimulating factor (GM-CSF) have been shown to be beneficial in reducing duration of systemic antibiotic therapy and in-patient hospitalization by decreasing the period of neutropenia in cancer patients undergoing chemotherapy. Since the underlying mechanism is unclear, the aim of this study was to investigate whether the administration of G-CSF and GM-CSF in two different doses (low dose and standard dose) would result into resolution of neutropenia with concomitant increase in multiple forms of dihydrofolate reductase (DHFR, a pivotal enzyme in the pathway of de novo DNA synthesis). Thirty seven cancer patients (26 males and 11 females; age 14-73 years) having chemotherapy-induced neutropenia (absolute neutrophil counts <500/µl) were treated with colony stimulating factor (CSF) in the following manner: 11 received GM-CSF (7 received a dose 250 µg/m2 and 4 received a dose of 100 µg/m(2)); 26 received G-CSF (14 received a dose of 5 µg/kg and 12 received a dose of 2.5 µg/kg). CSFs was given every day till the absolute neutrophil count was more than 1,000/µl. Ten ml blood was collected from each patient and analyzed for total leukocyte count (TLC) and active DHFR and immunoreactive nonfunctional form of DHFR (IRE) in the cytoplasm of blood leukocytes by using methotrexate binding assay and enzyme-linked immunosorbent assay (ELISA). A significant increase (p<0.05) in concentrations of both active DHFR and IRE following stimulation with low as well as standard doses of CSFs was observed along with increase in the TLC. There was no significant difference in number of days to resolution of neutropenia at these two doses, indicating that even low doses of CSFs are clinically effective. Along with an increase in TLC, the levels of DHFR increased even at low doses of CSF suggesting that this might be one of the mechanisms for CSF-induced proliferation of leukocytes in neutropenic cancer patients.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Neoplasias/sangre , Neutropenia/tratamiento farmacológico , Neutropenia/enzimología , Tetrahidrofolato Deshidrogenasa/metabolismo , Adolescente , Adulto , Anciano , Antineoplásicos/efectos adversos , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Recuento de Leucocitos , Leucocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Estudios Prospectivos , Adulto Joven
19.
Am J Reprod Immunol ; 67(4): 334-40, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22380628

RESUMEN

Recurrent spontaneous abortion (RSA), three or more pregnancy losses prior to 20 weeks, occurs in about 1% of all pregnancies, 50% of RSA cases remain unexplained and unresolved. Recently, immune pathways have been implicated in the pathophysiology of RSA. Immune tolerance of the fetal-placental unit and placental angiogenesis are mandatory for a successful pregnancy outcome. Unscheduled dysregulation of the placental vasculature is thought to be the pathophysiologic mechanisms underlying an array of pregnancy complications like infertility, miscarriage, pre-eclampsia, and fetal growth restriction and death. Investigations on mechanisms and management of RSA are mired by substandard design and lack of optimal randomized clinical trials and have resulted in disagreement on guidelines for evaluation and treatments for patients with multiple pregnancy losses of unknown etiology. The present review focuses on evidence-based research discussion with immunologic causes, and immune-regulatory therapies recommended for helping patients with a history of RSA. We highlight data that might support revalidation of low molecular weight heparin as a protective therapy in RSA. Newly launched growth factors, GM-CSF, and potentially novel agents to suppress inflammatory rejection, including regulatory T cells, human chorionic gonadotropin, and M-CSF/IL-10, may work in concert with tender-loving-care therapy and give hope to couples with multiple pregnancy losses.


Asunto(s)
Aborto Habitual/inmunología , Aborto Habitual/terapia , Gonadotropina Coriónica/uso terapéutico , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Heparina de Bajo-Peso-Molecular/uso terapéutico , Humanos , Recién Nacido , Interleucina-10/uso terapéutico , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Embarazo , Resultado del Embarazo , Linfocitos T Reguladores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA