Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
J Biochem Mol Toxicol ; 37(12): e23487, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37718545

RESUMO

Resistance to chemotherapy in cancer leads to poor therapeutic outcomes and also leads to challenges in treatment. The present work evaluated the mechanism involved in the resistance of 5-flurouracil (5-FU) in pancreatic cancer. At least 14 different pancreatic cancer (PC) cell lines were used for the study. For in vivo study female nude mice were selected. Patient-derived tumor xenograft samples were obtained from patients. The study involved, study for glucose uptake, fluorescence-activated cell sorting for glucose transporter, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide for cell survival, Picto-micrography for clonogenic assay, glutamine uptake assay, extracellular acidification and oxygen consumption rate, carbon dioxide release assay and lactate assay were also done. In addition to this, quantitative real-time polymerase chain reaction analysis for expression of genes, chromatin immunoprecipitation assay, western blot for protein expression, and immunohistochemical analysis in tumor sections, the tumors were studied by imaging for hypoxia and localization of TKT and CTPS-2. Also, patient-derived xenograft tumors were engrafted in nude mice, followed by a glucose uptake assay. We reported that elevated glycolytic flux causes dependence on glucose in cancer cells and, at the same time, increases pyrimidine biosynthesis. It was also found that stem cell factor-mediated stabilization of hypoxia-inducible factor-1a (HIF-1α) modulates the resistance in PC. Targeting HIF-1α in combination with 5-FU, strongly reduced the tumor burden. The study concludes that stem cell factor modulates HIF-1α and decreases the sensitivity in 5-FU resistant pancreatic cancer cells by targeting glucose metabolism. Deceased expression levels of CTPS-2 and TKT, which are regulators of pyrimidine biosynthesis could better the chance of survival in patients of pancreatic cancer receiving treatment of 5-FU.


Assuntos
Neoplasias Pancreáticas , Fator de Células-Tronco , Animais , Camundongos , Humanos , Feminino , Camundongos Nus , Fator de Células-Tronco/uso terapêutico , Neoplasias Pancreáticas/metabolismo , Glucose/metabolismo , Hipóxia , Linhagem Celular Tumoral , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Pirimidinas , Subunidade alfa do Fator 1 Induzível por Hipóxia
2.
Oncogene ; 42(34): 2578-2588, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37468679

RESUMO

Gastrointestinal stromal tumor (GIST) is the most common human sarcoma and is typically driven by a single mutation in the Kit or PDGFRA receptor. While highly effective, tyrosine kinase inhibitors (TKIs) are not curative. The natural ligand for the Kit receptor is Kit ligand (KitL), which exists in both soluble and membrane-bound forms. While KitL is known to stimulate human GIST cell lines in vitro, we used a genetically engineered mouse model of GIST containing a common human KIT mutation to investigate the intratumoral sources of KitL, importance of KitL during GIST oncogenesis, and contribution of soluble KitL to tumor growth in vivo. We discovered that in addition to tumor cells, endothelia and smooth muscle cells produced KitL in KitV558Δ/+ tumors, even after imatinib therapy. Genetic reduction of total KitL in tumor cells of KitV558Δ/+ mice impaired tumor growth in vivo. Similarly, genetic reduction of tumor cell soluble KitL in KitV558Δ/+ mice decreased tumor size. By RNA sequencing, quantitative PCR, and immunohistochemistry, KitL expression was heterogeneous in human GIST specimens. In particular, PDGFRA-mutant tumors had much higher KitL expression than Kit-mutant tumors, suggesting the benefit of Kit activation in the absence of mutant KIT. Serum KitL was higher in GIST patients with tumors resistant to imatinib and in those with tumors expressing more KitL RNA. Overall, KitL supports the growth of GIST at baseline and after imatinib therapy and remains a potential biomarker and therapeutic target.


Assuntos
Antineoplásicos , Tumores do Estroma Gastrointestinal , Humanos , Camundongos , Animais , Mesilato de Imatinib/farmacologia , Mesilato de Imatinib/uso terapêutico , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/patologia , Fator de Células-Tronco/genética , Fator de Células-Tronco/farmacologia , Fator de Células-Tronco/uso terapêutico , Pirimidinas/farmacologia , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Benzamidas/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas Proto-Oncogênicas c-kit , Mutação , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico
3.
Cells ; 12(5)2023 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-36899841

RESUMO

Severe traumatic brain injury (TBI) causes long-term disability and death in young adults. White matter is vulnerable to TBI damage. Demyelination is a major pathological change of white matter injury after TBI. Demyelination, which is characterized by myelin sheath disruption and oligodendrocyte cell death, leads to long-term neurological function deficits. Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) treatments have shown neuroprotective and neurorestorative effects in the subacute and chronic phases of experimental TBI. Our previous study has revealed that combined SCF and G-CSF treatment (SCF + G-CSF) enhances myelin repair in the chronic phase of TBI. However, the long-term effect and mechanism of SCF + G-CSF-enhanced myelin repair remain unclear. In this study, we uncovered persistent and progressive myelin loss in the chronic phase of severe TBI. SCF + G-CSF treatment in the chronic phase of severe TBI enhanced remyelination in the ipsilateral external capsule and striatum. The SCF + G-CSF-enhanced myelin repair is positively correlated with the proliferation of oligodendrocyte progenitor cells in the subventricular zone. These findings reveal the therapeutic potential of SCF + G-CSF in myelin repair in the chronic phase of severe TBI and shed light on the mechanism underlying SCF + G-CSF-enhanced remyelination in chronic TBI.


Assuntos
Lesões Encefálicas Traumáticas , Doenças Desmielinizantes , Remielinização , Humanos , Fator de Células-Tronco/metabolismo , Fator de Células-Tronco/uso terapêutico , Lesões Encefálicas Traumáticas/patologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Doenças Desmielinizantes/tratamento farmacológico
4.
Indian J Dermatol Venereol Leprol ; 88(6): 761-770, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35389028

RESUMO

Background Melasma is an acquired dyschromia with several histologic alterations in the epidermis, basement membrane and upper dermis. The treatment of melasma is challenging due to the irregular response and chronicity of the disease. To date, there are no curative strategies, largely due to the limited understanding of the intrinsic effects of each treatment. Objectives The objective of the study was to evaluate the histological changes promoted by triple combination cream, with or without complementary treatment with microneedling and oral tranexamic acid, in the treatment of melasma. Methods A factorial, randomised, controlled and evaluator-blinded clinical trial was performed involving 64 women with facial melasma, divided in four groups, who underwent 60 days of treatment with triple combination cream alone (control group) or combined with two monthly microneedling sessions (microneedling group), TA 250 mg twice daily (tranexamic acid group), or both tranexamic acid group and microneedling group. The participants underwent biopsy of the area with melasma at inclusion (D1) and D60. The primary outcomes were the variation (D1 × D60) between the variables: Thickness of the epidermis and stratum corneum, stratum corneum compaction and solar elastosis; melanin density in the epidermis and upper dermis; proportion between the extension of the nonintact and intact basement membrane zone; mast cell count in the upper dermis; melanocyte count in the basal layer, pendulum melanocyte count and melanocyte area; immunostaining density of vascular endothelial growth factor; stem cell factor and keratinocyte growth factor. Results One participant in the TG discontinued tranexamic acid due persistent headache; and herpes simplex occurred in three patients after microneedling. The groups showed a 24% (CI95%: 17-35%; P < 0.01) reduction in epidermal melanin density. There was no change in dermal melanin density or the area of melanocytes after treatment. There was an overall 25% (CI95%: 7-42%; P < 0.01) reduction in the number of pendulum melanocytes, especially in the microneedling and tranexamic acid group, that presented a 41% (CI95%: 7-73%; P < 0.01) reduction. The extension of the nonintact basal membrane relative to the intact basal membrane decreased after treatment, especially in microneedling group and microneedling and tranexamic acid group. There was an increase of 13% (CI95%: 5-21%; P = 0.02) in epidermal thickness and 6% (CI95%: 0-22%; P = 0.04) thinning of the stratum corneum in the groups. All groups showed stratum corneum compaction. Solar elastosis improved only in the microneedling group and microneedling and tranexamic acid group. Vascular endothelial growth factor immunostaining increased 14% (CI95%: 4-24%; P = 0.03) in the groups; and stem cell factor increased only in microneedling group. There was no change in the number of mast cells, CD34 and keratinocyte growth factor immunostaining. Limitations The site of biopsy may not represent all of the facial melasma and the immunohistochemical sensitivity of the cytokines does not have a stoichiometric relationship with proteins. Conclusion A greater thickness of the epidermis is associated with melasma bleaching. Dermal melanin seems to have no impact on melasma prognosis. Damage to the skin barrier and stimulus of angiogenesis should be avoided in the treatment of melasma. Microneedling complements the topical treatment of melasma by improving patterns of skin photoaging. Oral tranexamic acid complements the topical treatment of melasma by inhibiting the stem cell factor.


Assuntos
Melanose , Ácido Tranexâmico , Humanos , Feminino , Fator 7 de Crescimento de Fibroblastos/uso terapêutico , Melaninas , Fator A de Crescimento do Endotélio Vascular , Fator de Células-Tronco/uso terapêutico , Melanose/terapia , Melanose/tratamento farmacológico , Resultado do Tratamento
5.
Cell Transplant ; 27(4): 637-647, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29871518

RESUMO

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leucoencephalopathy (CADASIL) is a cerebral small vascular disease caused by NOTCH3 mutation-induced vascular smooth muscle cell (VSMC) degeneration, leading to ischemic stroke and vascular dementia. Our previous study has demonstrated that repeated treatment with a combination of stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) reduces VSMC degeneration and cerebral endothelial cell (EC) damage and improves cognitive function in a mouse model of CADASIL (TgNotch3R90C). This study aimed to determine whether cerebral thrombosis occurs in TgNotch3R90C mice and whether repeated SCF+G-CSF treatment reduces cerebral thrombosis in TgNotch3R90C mice. Using the approaches of bone marrow transplantation to track bone marrow-derived cells and confocal imaging, we observed bone marrow-derived blood cell occlusion in cerebral small vessels and capillaries (thrombosis). Most thrombosis occurred in the cerebral capillaries (93% of total occluded vessels), and the thrombosis showed an increased frequency in the regions of capillary bifurcation. Degenerated capillary ECs were seen inside and surrounding the thrombosis, and the bone marrow-derived ECs were also found next to the thrombosis. IgG extravasation was seen in and next to the areas of thrombosis. SCF+G-CSF treatment significantly reduced cerebral capillary thrombosis and IgG extravasation. These data suggest that the EC damage is associated with thrombosis and blood-brain barrier leakage in the cerebral capillaries under the CADASIL-like condition, whereas SCF+G-CSF treatment diminishes these pathological alterations. This study provides new insight into the involvement of cerebral capillary thrombosis in the development of CADASIL and potential approaches to reduce the thrombosis, which may restrict the pathological progression of CADASIL.


Assuntos
CADASIL/tratamento farmacológico , Capilares/patologia , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Trombose Intracraniana/tratamento farmacológico , Fator de Células-Tronco/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Encéfalo/patologia , CADASIL/complicações , CADASIL/patologia , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoglobulina G/metabolismo , Trombose Intracraniana/complicações , Trombose Intracraniana/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Notch/metabolismo , Fator de Células-Tronco/farmacologia
6.
J Neurosurg ; 129(5): 1286-1294, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29372883

RESUMO

OBJECTIVETraumatic brain injury (TBI) is a major cause of long-term disability and death in young adults. The lack of pharmaceutical therapy for post-acute TBI recovery remains a crucial medical challenge. Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF), which are 2 key hematopoietic growth factors, have shown neuroprotective and neurorestorative effects in experimental stroke. The objective of this study was to determine the therapeutic efficacy of combined treatment (SCF + G-CSF) in subacute TBI.METHODSYoung-adult male C57BL mice were subject to TBI in the cortex of the right hemisphere. After TBI induction, mice were randomly divided into 2 groups: a vehicle control group and an SCF + G-CSF treatment group. Mice without TBI served as sham operative controls. Treatment was initiated 2 weeks after TBI induction. SCF (200 µg/kg) and G-CSF (50 µg/kg) or an equal volume of vehicle solution was subcutaneously injected daily for 7 days. A battery of neurobehavioral tests for evaluation of memory and cognitive function (water maze and novel object recognition tests), anxiety (elevated plus maze test), and motor function (Rota-Rod test) was performed during the period of 2-9 weeks after treatment. Neurodegeneration and dendritic density in both hemispheres were determined through histochemistry and immunohistochemistry at 11 weeks posttreatment.RESULTSWater maze testing showed that TBI-impaired spatial learning and memory was restored by SCF + G-CSF treatment. The findings from the elevated plus maze tests revealed that SCF + G-CSF treatment recovered TBI-caused anxiety and risk-taking behavior. There were no significant differences between the treated and nontreated TBI mice in both the Rota-Rod test and novel object recognition test. In the brain sections, the authors observed that widespread degenerating neurons were significantly increased in both hemispheres in the TBI-vehicle control mice. TBI-induced increases in neurodegeneration were significantly reduced by SCF + G-CSF treatment in the contralateral hemisphere, making it no different from that of the sham controls. Dendritic density in the frontal cortex of the contralateral hemisphere was significantly reduced in the TBI-vehicle control mice, whereas SCF + G-CSF-treated TBI mice showed significant increases of the dendritic density in the same brain region. SCF + G-CSF-treated TBI mice also showed a trend toward increasing dendritic density in the contralateral hippocampus.CONCLUSIONSSCF + G-CSF treatment in the subacute phase of TBI restored TBI-impaired spatial learning and memory, prevented posttraumatic anxiety and risk-taking behavior, inhibited TBI-induced neurodegeneration, and enhanced neural network remodeling. These findings suggest the therapeutic potential of hematopoietic growth factors for brain repair in the subacute phase of TBI.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Recuperação de Função Fisiológica/efeitos dos fármacos , Fator de Células-Tronco/uso terapêutico , Animais , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Reconhecimento Psicológico/efeitos dos fármacos , Aprendizagem Espacial/efeitos dos fármacos , Memória Espacial/efeitos dos fármacos , Fator de Células-Tronco/farmacologia , Resultado do Tratamento
7.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 25(5): 1544-1549, 2017 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-29070141

RESUMO

OBJECTIVE: To study the therapeutic effect of rhSCF early administration on rhesus monkeys with severe acute radiation sickness(ARS). METHODS: Twelve adult monkeys totally exposed to 7.0 Gy 60Co were divided into radiation control and SCF groups, and monkeys in SCF group were subcutaneously injected recombinant human SCF(rhSCF) 200 µg/kg at half an hour and 24 hour after irradiation, while the radiation control monkeys were injected physiological saline. Survival was monitored and hematopoiesis was evaluated at 40 days following early treatment. RESULTS: 6 animals treated with rhSCF all survived, while 2 in irradiated controls survived on 40 day after radiation. rhSCF treatment promoted hematopoiesis recovery significantly, increased the nadir of white blood cells, neutrophils and platelets, and simplified supportive care in ARS rhesus monkeys. CONCLUSION: RhSCF injection soon after TBI taken shows an significant therapeutic efficiency on rhesus monkeys with severe acute radiation sickness.


Assuntos
Lesões por Radiação/terapia , Proteínas Recombinantes/uso terapêutico , Fator de Células-Tronco/uso terapêutico , Animais , Hematopoese , Humanos , Macaca mulatta , Irradiação Corporal Total
8.
Medisan ; 21(5)mayo 2017.
Artigo em Espanhol | LILACS | ID: biblio-841702

RESUMO

Los avances en la medicina regenerativa han sido evidentes en los últimos años y esto se ha obtenido por los nuevos conocimientos alcanzados en relación con las células madre, cuyo uso en la terapia de reemplazo ha dado lugar a una nueva era en la medicina. A tales efectos se realizó una revisión bibliográfica con el objetivo de difundir sus generalidades y aplicaciones, así como lo relacionado con las investigaciones básicas que se realizan en ese campo y los principales logros obtenidos. La posibilidad de expansión y diferenciación de dichas células, permite obtener un número suficiente de estas, lo cual ayuda al desarrollo de la terapia celular


Advances in regenerative medicine have been evident in the last years and this has been obtained due to the new knowledge related to stem cells, which use in the replacement therapy has given rise to a new age in medicine. To such effects, a literature review was carried out aimed at spreading its generalities and implementations, as well as everything related to the basic investigations carried out in this field and the main achievements obtained. The possibility of expansion and differentiation of such cells, allow to obtain enough quantitiy of them, which helps the development of cell therapy


Assuntos
Humanos , Masculino , Feminino , Células-Tronco , Medicina Regenerativa , Terapia Baseada em Transplante de Células e Tecidos , Fator de Células-Tronco/uso terapêutico
9.
Neurobiol Dis ; 73: 189-203, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25251607

RESUMO

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a Notch3 dominant mutation-induced cerebral small vascular disease, is characterized by progressive degeneration of vascular smooth muscle cells (vSMCs) of small arteries in the brain, leading to recurrent ischemic stroke, vascular dementia and death. To date, no treatment can stop or delay the progression of this disease. Herein, we determined the therapeutic effects of stem cell factor (SCF) in combination with granulocyte colony-stimulating factor (G-CSF) (SCF+G-CSF) in a mouse model of CADASIL carrying the human mutant Notch3 gene. SCF+G-CSF was subcutaneously administered for 5 days and repeated 4 times with 1-4 month intervals. We found through water maze testing that SCF+G-CSF treatment improved cognitive function. SCF+G-CSF also attenuated vSMC degeneration in small arteries, increased cerebral blood vascular density, and inhibited apoptosis in CADASIL mice. We also discovered that loss of cerebral capillary endothelial cells and neural stem cells/neural progenitor cells (NSCs/NPCs) occurred in CADASIL mice. SCF+G-CSF treatment inhibited the CADASIL-induced cell loss in the endothelia and NSCs/NPCs and promoted neurogenesis. In an in vitro model of apoptosis, SCF+G-CSF prevented apoptotic cell death in vSMCs through AKT signaling and by inhibiting caspase-3 activity. These data suggest that SCF+G-CSF restricts the pathological progression of CADASIL. This study offers new insights into developing therapeutic strategies for CADASIL.


Assuntos
CADASIL/complicações , CADASIL/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Fator de Células-Tronco/uso terapêutico , Animais , Transplante de Medula Óssea , CADASIL/genética , CADASIL/cirurgia , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Mutação/genética , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Receptor Notch3 , Receptores Notch/genética , Fatores de Tempo
10.
PLoS One ; 9(10): e107891, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25309991

RESUMO

BACKGROUND: The gastrointestinal (GI) mucosal cells turnover regularly under physiological conditions, which may be stimulated in various pathological situations including inflammation. Local epithelial stem cells appear to play a major role in such mucosal renewal or pathological regeneration. Less is clear about the involvement of multipotent stem cells from blood in GI repair. We attempted to explore a role of bone marrow mesenchymal stromal cells (BMMSCs) and soluble stem cell factor (SCF) in GI mucosa regeneration in a rat model of inflammatory bowel diseases (IBD). METHODS: BMMSCs labelled with the fluorescent dye PKH26 from donor rats were transfused into rats suffering indomethacin-induced GI injury. Experimental effects by BMMSCs transplant and SCF were determined by morphometry of intestinal mucosa, double labeling of PKH26 positive BMMSCs with endogenous proliferative and intestinal cell markers, and western blot and PCR analyses of the above molecular markers in the recipient rats relative to controls. RESULTS: PKH26 positive BMMSCs were found in the recipient mucosa, partially colocalizing with the proliferating cell nuclear antigen (PCNA), Lgr5, Musashi-1 and ephrin-B3. mRNA and protein levels of PCNA, Lgr5, Musashi-1 and ephrin-B3 were elevated in the intestine in BMMSCs-treated rats, most prominent in the BMMSCs-SCF co-treatment group. The mucosal layer and the crypt layer of the small intestine were thicker in BMMSCs-treated rats, more evident in the BMMSCs-SCF co-treatment group. CONCLUSION: BMMSCs and SCF participate in but may play a synergistic role in mucosal cell regeneration following experimentally induced intestinal injury. Bone marrow stem cell therapy and SCF administration may be of therapeutic value in IBD.


Assuntos
Células da Medula Óssea , Doenças Inflamatórias Intestinais/terapia , Mucosa Intestinal/fisiologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Regeneração/fisiologia , Fator de Células-Tronco/uso terapêutico , Animais , Modelos Animais de Doenças , Indometacina , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
11.
J Neurosci Res ; 92(7): 856-69, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24936617

RESUMO

Amyotrophic lateral sclerosis (ALS) is a progressive disease associated with motor neuron death. Several experimental treatments, including cell therapy using hematopoietic or neuronal stem cells, have been tested in ALS animal models, but therapeutic benefits have been modest. Here we used a new therapeutic strategy, bone marrow transplantation (BMT) with stem cell factor (SCF)- or FMS-like tyrosine kinase 3 (flt3)-activated bone marrow (BM) cells for the treatment of hSOD1(G93A) transgenic mice. Motor function and survival showed greater improvement in the SCF group than in the group receiving BM cells that had not been activated (BMT alone group), although no improvement was shown in the flt3 group. In addition, larger numbers of BM-derived cells that expressed the microglia marker Iba1 migrated to the spinal cords of recipient mice compared with the BMT alone group. Moreover, after SCF activation, but not flt3 activation or no activation, the migrating microglia expressed glutamate transporter-1 (GLT-1). In spinal cords in the SCF group, inflammatory cytokines tumor necrosis factor-α and interleukin-1ß were suppressed and the neuroprotective molecule insulin-like growth factor-1 increased relative to nontreatment hSOD1(G93A) transgenic mice. Therefore, SCF activation changed the character of the migrating donor BM cells, which resulted in neuroprotective effects. These studies have identified SCF-activated BM cells as a potential new therapeutic agent for the treatment of ALS.


Assuntos
Esclerose Lateral Amiotrófica/cirurgia , Transplante de Medula Óssea/métodos , Movimento Celular/fisiologia , Microglia/fisiologia , Fator de Células-Tronco/uso terapêutico , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Movimento Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios Motores/fisiologia , Teste de Desempenho do Rota-Rod , Medula Espinal/metabolismo , Medula Espinal/patologia , Superóxido Dismutase/genética , Tirosina Quinase 3 Semelhante a fms/uso terapêutico
12.
Molecules ; 19(4): 4770-8, 2014 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-24743934

RESUMO

This article concisely summarizes data on the action of one of the principal and best known growth factors, the granulocyte colony-stimulating factor (G-CSF), in a mammalian organism exposed to radiation doses inducing acute radiation syndrome. Highlighted are the topics of its real or anticipated use in radiation accident victims, the timing of its administration, the possibilities of combining G-CSF with other drugs, the ability of other agents to stimulate endogenous G-CSF production, as well as of the capability of this growth factor to ameliorate not only the bone marrow radiation syndrome but also the gastrointestinal radiation syndrome. G-CSF is one of the pivotal drugs in the treatment of radiation accident victims and its employment in this indication can be expected to remain or even grow in the future.


Assuntos
Síndrome Aguda da Radiação/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Síndrome Aguda da Radiação/patologia , Animais , Medula Óssea/patologia , Medula Óssea/efeitos da radiação , Esquema de Medicação , Quimioterapia Combinada , Fator Estimulador de Colônias de Granulócitos/biossíntese , Humanos , Interleucina-3/uso terapêutico , Proteínas de Membrana/uso terapêutico , Liberação Nociva de Radioativos , Proteínas Recombinantes/uso terapêutico , Fator de Células-Tronco/uso terapêutico , Trombopoetina/uso terapêutico , Fatores de Tempo
13.
Circ Res ; 114(1): 41-55, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24170267

RESUMO

RATIONALE: Hypoxia favors stem cell quiescence, whereas normoxia is required for stem cell activation, but whether cardiac stem cell (CSC) function is regulated by the hypoxic/normoxic state of the cell is currently unknown. OBJECTIVE: A balance between hypoxic and normoxic CSCs may be present in the young heart, although this homeostatic control may be disrupted with aging. Defects in tissue oxygenation occur in the old myocardium, and this phenomenon may expand the pool of hypoxic CSCs, which are no longer involved in myocyte renewal. METHODS AND RESULTS: Here, we show that the senescent heart is characterized by an increased number of quiescent CSCs with intact telomeres that cannot re-enter the cell cycle and form a differentiated progeny. Conversely, myocyte replacement is controlled only by frequently dividing CSCs with shortened telomeres; these CSCs generate a myocyte population that is chronologically young but phenotypically old. Telomere dysfunction dictates their actual age and mechanical behavior. However, the residual subset of quiescent young CSCs can be stimulated in situ by stem cell factor reversing the aging myopathy. CONCLUSIONS: Our findings support the notion that strategies targeting CSC activation and growth interfere with the manifestations of myocardial aging in an animal model. Although caution has to be exercised in the translation of animal studies to human beings, our data strongly suggest that a pool of functionally competent CSCs persists in the senescent heart and that this stem cell compartment can promote myocyte regeneration effectively, partly correcting the aging myopathy.


Assuntos
Envelhecimento/efeitos dos fármacos , Cardiomiopatias/metabolismo , Hipóxia/metabolismo , Mioblastos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Células-Tronco/farmacologia , Nicho de Células-Tronco , Envelhecimento/metabolismo , Animais , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/patologia , Ciclo Celular , Linhagem da Célula , Proliferação de Células , Senescência Celular/efeitos dos fármacos , Hipóxia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos Cardíacos/efeitos dos fármacos , Mioblastos Cardíacos/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Fator de Células-Tronco/uso terapêutico , Homeostase do Telômero
14.
Pediatr Res ; 74(6): 682-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24153399

RESUMO

BACKGROUND: Stem cell factor (SCF) and its receptor, c-kit, are modulators of angiogenesis. Neonatal hyperoxia-induced lung injury (HILI) is characterized by disordered angiogenesis. The objective of this study was to determine whether exogenous SCF improves recovery from neonatal HILI by improving angiogenesis. METHODS: Newborn rats assigned to normoxia (RA: 20.9% O2) or hyperoxia (90% O2) from postnatal day (P) 2 to 15, received daily injections of SCF 100 µg/kg or placebo (PL) from P15 to P21. Lung morphometry was performed at P28. Capillary tube formation in SCF-treated hyperoxia-exposed pulmonary microvascular endothelial cells (HPMECs) was determined by Matrigel assay. RESULTS: As compared with RA, hyperoxic-PL pups had decrease in alveolarization and in lung vascular density, and this was associated with increased right ventricular systolic pressure (RVSP), right ventricular hypertrophy, and vascular remodeling. In contrast, SCF-treated hyperoxic pups had increased angiogenesis, improved alveolarization, and attenuation of pulmonary hypertension as evidenced by decreased RVSP, right ventricular hypertrophy, and vascular remodeling. Moreover, in an in vitro model, SCF increased capillary tube formation in hyperoxia-exposed HPMECs. CONCLUSION: Exogenous SCF restores alveolar and vascular structure in neonatal rats with HILI by promoting neoangiogenesis. These findings suggest a new strategy to treat lung diseases characterized by dysangiogenesis.


Assuntos
Hiperóxia/tratamento farmacológico , Lesão Pulmonar/tratamento farmacológico , Fator de Células-Tronco/uso terapêutico , Animais , Animais Recém-Nascidos , Hiperóxia/fisiopatologia , Lesão Pulmonar/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Placebos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/crescimento & desenvolvimento , Ratos , Ratos Sprague-Dawley , Fator de Células-Tronco/farmacologia
15.
Recent Pat CNS Drug Discov ; 8(1): 2-12, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23173646

RESUMO

Stroke is a serious cerebrovascular disease that causes high mortality and persistent disability in adults worldwide. Stroke is also an enormous public health problem and a heavy public financial burden in the United States. Treatment for stroke is very limited. Thrombolytic therapy by tissue plasminogen activator (tPA) is the only approved treatment for acute stroke, and no effective treatment is available for chronic stroke. Developing new therapeutic strategies, therefore, is a critical need for stroke treatment. This article summarizes the discovery of new routes of treatment for acute and chronic stroke using two hematopoietic growth factors, stem cell factor (SCF) and granulocyte-colony stimulating factor (G-CSF). In a study of acute stroke, SCF and G-CSF alone or in combination displays neuroprotective effects in an animal model of stroke. SCF appears to be the optimal treatment for acute stroke as the functional outcome is superior to G-CSF alone or in combination (SCF+G-CSF); however, SCF+G-CSF does show better functional recovery than G-CSF. In a chronic stroke study, the therapeutic effects of SCF and G-CSF alone or in combination appear differently as compared with their effects on the acute stroke. SCF+G-CSF induces stable and long-lasting functional improvement; SCF alone also improves functional outcome but its effectiveness is less than SCF+G-CSF, whereas G-CSF shows no therapeutic effects. Although the mechanism by which SCF+G-CSF repairs the brain in chronic stroke remains poorly understood, our recent findings suggest that the SCF+G-CSF-induced functional improvement in chronic stroke is associated with a contribution to increasing angiogenesis and neurogenesis through bone marrow-derived cells and the direct effects on stimulating neurons to form new neuronal networks. These findings would assist in developing new treatment for stroke. The article presents some promising patents on role of stem cell factor and granulocyte-colony stimulating factor in treatment of stroke.


Assuntos
Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Fator de Células-Tronco/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Indutores da Angiogênese/uso terapêutico , Animais , Doença Crônica , Quimioterapia Combinada , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Humanos , Neurogênese/efeitos dos fármacos , Recuperação de Função Fisiológica/efeitos dos fármacos , Fator de Células-Tronco/administração & dosagem
16.
Exp Mol Pathol ; 94(1): 247-54, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22750282

RESUMO

OBJECTIVES: Osteonecrosis is also known as avascular necrosis, and two types of cell death are included in the pathogenesis of osteonecrosis: necrosis and apoptosis. Apoptosis in the osteonecrosis of femoral head is thought to be the key determinant of glucocorticoid-induced cortical bone loss. The present study was implemented to evaluate the anti-apoptotic effect of Granulocyte colony-stimulating factor and stem cell factor (G-CSF/SCF) in rabbits with steroid-induced osteonecrosis. METHODS: In the experiment, osteonecrosis was induced by low-dose lipopolysaccharide and subsequent pulsed high-dose methylprednisolone. Rabbits in preventive medicine group were treated with 100 µg/kg/d G-CSF and 25 µg/kg/d SCF. Then hematological and histomorphometric methods were used to investigate the treatment effects of osteonecrosis. Apoptosis was assessed via quantitative TUNEL staining and activated caspase-3 immunostaining and immunoblotting. RESULTS: The results showed that G-CSF/SCF treatment could increase the secretion of serum osteocalcin, but inhibit the expression of serum tartrate-resistant acid phosphatase (TRAP5b). The incidence of osteonecrosis was significantly decreased in Preventive group when compared with Steroid group (42.1% vs. 88.2%). Histomorphometric analysis showed that G-CSF/SCF pre-disposal treatment was able to increase trabecular mineral appositional rate (MAR) and bone formation rate (BFR). Quantitative TUNEL and activated caspase-3 levels showed lower apoptosis in the Preventive group. CONCLUSIONS: In conclusion, G-CSF/SCF treatment could inhibit caspase-3-dependent apoptosis in osteocytes to exert beneficial effects in preventing steroid-induced ON in rabbit models.


Assuntos
Apoptose/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Osteonecrose/tratamento farmacológico , Osteonecrose/prevenção & controle , Fator de Células-Tronco/farmacologia , Fosfatase Ácida/sangue , Animais , Caspase 3/sangue , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Isoenzimas/sangue , Metilprednisolona , Osteocalcina/sangue , Osteogênese/efeitos dos fármacos , Osteonecrose/induzido quimicamente , Coelhos , Fator de Células-Tronco/uso terapêutico , Fosfatase Ácida Resistente a Tartarato
17.
Eur Cytokine Netw ; 23(2): 56-63, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22668525

RESUMO

We have re-evaluated the benefit of using erythropoietin (Epo) as a pleiotropic cytokine to counteract hematological and extra-hematological toxicity following lethal irradiation. B6D2F1 mice were exposed to a dose of 9 Gy gamma radiation resulting in 90% mortality at 30 days, and then injected with stem cell factor, FLT-3 ligand, thrombopoietin and interleukin-3 [i.e. SFT3] at two and 24 hours with or without Epo (1,000 IU/kg) at 2 hours and day 8. As controls, two groups of irradiated mice were given only Epo or Phosphate-buffered saline. Epo synergized with SFT3 to rescue lethally-irradiated mice from radiation-induced death (survival: 60%, 95% and 5% respectively for SFT3, SFT3+Epo and controls at 30 days, p<0.05), whereas Epo alone exhibited no protective effect. Hematopoietic parameters did not differ significantly between SFT3 and SFT3+Epo groups during the animal death period. Some beneficial effects on gastro-intestinal toxicity were noticed following administration of Epo, although lung, liver and kidney were not protected. Further studies are necessary to understand fully the mechanisms involved in these effects of Epo in order to optimize treatment with cytokines following high-dose irradiation.


Assuntos
Citocinas/uso terapêutico , Eritropoetina/uso terapêutico , Lesões por Radiação/tratamento farmacológico , Protetores contra Radiação/uso terapêutico , Fator de Células-Tronco/uso terapêutico , Animais , Citocinas/administração & dosagem , Sinergismo Farmacológico , Eritropoetina/administração & dosagem , Raios gama , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/efeitos da radiação , Interleucina-3/administração & dosagem , Interleucina-3/uso terapêutico , Rim/efeitos dos fármacos , Rim/efeitos da radiação , Fígado/efeitos dos fármacos , Fígado/efeitos da radiação , Pulmão/efeitos dos fármacos , Pulmão/efeitos da radiação , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/uso terapêutico , Camundongos , Protetores contra Radiação/administração & dosagem , Distribuição Aleatória , Fator de Células-Tronco/administração & dosagem , Trombopoetina/administração & dosagem , Trombopoetina/uso terapêutico , Irradiação Corporal Total
18.
Stem Cell Res Ther ; 3(1): 7, 2012 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-22356811

RESUMO

Avascular necrosis (AVN) of the femoral head is a debilitating disease of multifactorial genesis, predominately affects young patients, and often leads to the development of secondary osteoarthritis. The evolving field of regenerative medicine offers promising treatment strategies using cells, biomaterial scaffolds, and bioactive factors, which might improve clinical outcome. Early stages of AVN with preserved structural integrity of the subchondral plate are accessible to retrograde surgical procedures, such as core decompression to reduce the intraosseous pressure and to induce bone remodeling. The additive application of concentrated bone marrow aspirates, ex vivo expanded mesenchymal stem cells, and osteogenic or angiogenic growth factors (or both) holds great potential to improve bone regeneration. In contrast, advanced stages of AVN with collapsed subchondral bone require an osteochondral reconstruction to preserve the physiological joint function. Analogously to strategies for osteochondral reconstruction in the knee, anterograde surgical techniques, such as osteochondral transplantation (mosaicplasty), matrix-based autologous chondrocyte implantation, or the use of acellular scaffolds alone, might preserve joint function and reduce the need for hip replacement. This review summarizes recent experimental accomplishments and initial clinical findings in the field of regenerative medicine which apply cells, growth factors, and matrices to address the clinical problem of AVN.


Assuntos
Necrose da Cabeça do Fêmur/terapia , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Células-Tronco Mesenquimais/citologia , Proteínas Morfogenéticas Ósseas/uso terapêutico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Fator de Crescimento de Hepatócito/uso terapêutico , Humanos , Transplante de Células-Tronco Mesenquimais , Medicina Regenerativa , Fator de Células-Tronco/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
19.
Leuk Lymphoma ; 52(8): 1435-45, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21635205

RESUMO

In a substantial number of patients with aplastic anemia (AA), immunosuppressive therapy (IST) with antithymocyte globulin (ATG) and cyclosporine A (CSA) leads to long-lasting remissions and is thus regarded as standard therapy. However, no consensus exists on how to treat refractory or relapsed AA, especially when no related stem cell donor is available. For selected patients, matched unrelated donor stem cell transplant (MUDSCT) is an option. In addition, umbilical cord blood and haploidentical donors have been considered as an alternative source of stem cells. Patients without a suitable donor may benefit from a second cycle of ATG and CSA. Alternatives are alemtuzumab and high dose cyclophosphamide, both of which induce remission in more than 50% of patients with relapsed AA. Further experimental drugs are androgens, hematopoietic growth factors (interleukins IL-3, IL-6, and IL-11 and stem cell factor [SCF]), and the tumor necrosis factor (TNF)-targeting agent etanercept. Clinical trials with these agents are ongoing and will explore long-term outcomes and potential beneficial effects of drug combinations.


Assuntos
Anemia Aplástica/terapia , Imunossupressores/uso terapêutico , Transplante de Células-Tronco/métodos , Alemtuzumab , Androgênios/uso terapêutico , Anemia Aplástica/diagnóstico , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Resistência a Medicamentos , Humanos , Interleucinas/uso terapêutico , Recidiva , Fator de Células-Tronco/uso terapêutico
20.
Bone Marrow Transplant ; 46(7): 936-42, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20956952

RESUMO

Ancestim (r-MetHuSCF) is available in France for compassionate use in patients who are candidates for high-dose chemotherapy and autologous transplantation, and who failed in previous attempts at mobilization and collection. We report here data from 513 adult patients who benefited from this program, between January 1998 and July 2007. Given with systematic premedication, ancestim was generally well tolerated, although severe but not life-threatening adverse events were reported in 12 individuals. Overall, a graft was obtained or completed for 235 patients (46%). The median number of collected CD34+ cells was 3.00 × 10(6)/kg (range: 0.03-39.50). The target threshold of 2 × 10(6) CD34+ cells/kg was reached in 161 patients (31%). Factors associated with collection were diagnosis of myeloma, no previous autologous transplant, no more than one previous failed attempt and a mobilization regimen including cytotoxic agents. A total of 207 patients (40%) proceeded to high-dose chemotherapy and autologous transplantation. The median time to reach 0.5 × 10(9)/L neutrophils and 20 × 10(9)/L platelets was 12 (6-40) and 13 (0-31) days, respectively. We conclude that a combination of ancestim with filgrastim successfully mobilized CD34+ cells in peripheral blood, and allowed adequate collection in preparation for autologous transplantation in approximately one-third of poorly mobilizing patients.


Assuntos
Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Mobilização de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Neoplasias/patologia , Fator de Células-Tronco/análogos & derivados , Adolescente , Adulto , Ensaios de Uso Compassivo , Filgrastim , Fator Estimulador de Colônias de Granulócitos/efeitos adversos , Humanos , Pessoa de Meia-Idade , Neoplasias/sangue , Neoplasias/cirurgia , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/uso terapêutico , Fator de Células-Tronco/efeitos adversos , Fator de Células-Tronco/uso terapêutico , Transplante Autólogo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...