Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Stem Cells ; 34(8): 2145-56, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27090492

RESUMO

Hematopoietic stem/progenitor cell (HSPC) mobilization is an essential homeostatic process regulated by the interaction of cellular and molecular components in bone marrow niches. It has been shown by others that neurotransmitters released from the sympathetic nervous system regulate HSPC egress from bone marrow to peripheral blood. In this study, we investigate the functional role of neuropeptide Y (NPY) on this process. NPY deficient mice had significantly impaired HSPC mobilization due to increased expression of HSPC maintenance factors by reduction of matrix metalloproteinase-9 (MMP-9) activity in bone marrow. Pharmacological or endogenous elevation of NPY led to decrease of HSPC maintenance factors expression by activating MMP-9 in osteoblasts, resulting in HSPC mobilization. Mice in which the Y1 receptor was deleted in osteoblasts did not exhibit HSPC mobilization by NPY. Furthermore, NPY treatment in ovariectomized mice caused reduction of bone loss due to HSPC mobilization. These results suggest a new role of NPY on HSPC mobilization, as well as the potential therapeutic application of this neuropeptide for stem cell-based therapy. Stem Cells 2016;34:2145-2156.


Assuntos
Mobilização de Células-Tronco Hematopoéticas , Metaloproteinase 9 da Matriz/metabolismo , Neuropeptídeo Y/metabolismo , Osteoblastos/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Animais , Osso e Ossos/metabolismo , Quimiotaxia , Feminino , Homeostase , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/deficiência , Osteoblastos/citologia , Osteoblastos/enzimologia , Receptores CXCR4/metabolismo
2.
J Clin Neurol ; 20(3): 300-305, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38713076

RESUMO

BACKGROUND AND PURPOSE: Monoclonal antibodies (mAbs) targeting calcitonin-gene-related peptide (CGRP) or its receptor (anti-CGRP-R) have been widely administered to patients with migraine who show inadequate responses to preventive medications. Among patients in whom a particular anti-CGRP-R mAb is ineffective, switching between different anti-CGRP-R mAbs can be the next option. Few studies have investigated treatment outcomes for antibody switching, especially between mAbs with the same target of the CGRP ligand. We aimed to determine the treatment outcome after switching between two anti-CGRP mAbs (galcanezumab to fremanezumab). METHODS: We identified migraine patients in a prospective headache clinic registry who received galcanezumab for ≥3 months and were switched to fremanezumab for a further ≥3 months at a single university hospital. We defined a treatment response as a ≥50% reduction in the number of days with a moderate or severe headache at the third month of treatment relative to baseline. The treatment response after switching to fremanezumab was compared with the initial treatment response to galcanezumab. RESULTS: Among 21 patients identified in the registry, 7 (33.3%) were initial responders to galcanezumab. After switching to fremanezumab, 7 (33.3%) showed a treatment response. The treatment response rate was 28.6% in the initial responders and 71.4% in the nonresponders to galcanezumab (p>0.999). CONCLUSIONS: Switching between anti-CGRP mAbs (galcanezumab to fremanezumab) yielded a treatment outcome comparable to that reported previously when switching from an anti-CGRP-R mAb (erenumab) to an anti-CGRP mAb (galcanezumab or fremanezumab). The treatment response to fremanezumab seems to be independent of the prior treatment response to galcanezumab. Our findings suggest that switching to another anti-CGRP mAb can be considered when a particular anti-CGRP mAb is ineffective or intolerable.

3.
Sci Rep ; 13(1): 3773, 2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36882454

RESUMO

Increasing evidence suggests an association between SARS-CoV-2 vaccines and Guillain-Barré syndrome (GBS). Nevertheless, little is understood about the contributing risk factors and clinical characteristics of GBS post SARS-CoV-2 vaccination. In this prospective surveillance study of 38,828,691 SARS-CoV-2 vaccine doses administered from February 2021 to March 2022 in the Gyeonggi Province, South Korea, 55 cases of GBS were reported post vaccination. We estimated the incidence rate of GBS per million doses and the incidence rate ratio for the vaccine dose, mechanism, age, and sex. Additionally, we compared the clinical characteristics of GBS following mRNA-based and viral vector-based vaccinations. The overall incidence of GBS following SARS-CoV-2 vaccination was 1.42 per million doses. Viral vector-based vaccines were associated with a higher risk of GBS. Men were more likely to develop GBS than women. The third dose of vaccine was associated with a lower risk of developing GBS. Classic sensorimotor and pure motor subtypes were the predominant clinical subtypes, and demyelinating type was the predominant electrodiagnostic subtype. The initial dose of viral-vector based vaccine and later doses of mRNA-based vaccine were associated with GBS, respectively. GBS following SARS-CoV-2 vaccination may not be clinically distinct. However, physicians should pay close attention to the classic presentation of GBS in men receiving an initial dose of viral vector-based SARS-CoV-2 vaccines.


Assuntos
COVID-19 , Síndrome de Guillain-Barré , Vacinas Virais , Masculino , Humanos , Feminino , Incidência , Vacinas contra COVID-19/efeitos adversos , SARS-CoV-2 , Síndrome de Guillain-Barré/epidemiologia , Síndrome de Guillain-Barré/etiologia , Estudos Prospectivos , COVID-19/epidemiologia , COVID-19/prevenção & controle , Vacinação/efeitos adversos , RNA Mensageiro
5.
BMB Rep ; 50(3): 138-143, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27998395

RESUMO

Ovariectomy-induced bone loss is related to an increased deposition of osteoclasts on bone surfaces. We reported that the 36-amino-acid-long neuropeptide Y (NPY) could mobilize hematopoietic stem/progenitor cells (HSPCs) from the bone marrow to the peripheral blood by regulating HSPC maintenance factors and that mobilization of HSPCs ameliorated low bone density in an ovariectomy-induced osteoporosis mouse model by reducing the number of osteoclasts. Here, we demonstrated that new NPY peptides, recombined from the cleavage of the full-length NPY, showed better functionality for HSPC mobilization than the full-length peptide. These recombinant peptides mediated HSPC mobilization with greater efficiency by decreasing HSPC maintenance factors. Furthermore, treatment with these peptides reduced the number of osteoclasts and relieved ovariectomy-induced bone loss in mice more effectively than treatment with full-length NPY. Therefore, these results suggest that peptides recombined from full-length NPY can be used to treat osteoporosis. [BMB Reports 2017; 50(3): 138-143].


Assuntos
Mobilização de Células-Tronco Hematopoéticas/métodos , Neuropeptídeo Y/metabolismo , Neuropeptídeo Y/uso terapêutico , Osteoporose Pós-Menopausa/terapia , Animais , Medula Óssea/metabolismo , Células da Medula Óssea/metabolismo , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Osteoclastos/metabolismo , Osteoporose , Ovariectomia/efeitos adversos
6.
BMB Rep ; 49(5): 288-92, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26728272

RESUMO

Cisplatin is a platinum-based chemotherapeutic drug for treating various types of cancers. However, the use of cisplatin is limited by its negative effect on normal tissues, particularly nephrotoxicity. Various mechanisms such as DNA adduct formation, mitochondrial dysfunction, oxidative stress, and apoptosis are involved in the adverse effect induced by cisplatin treatment. Several studies have suggested that neuropeptide Y (NPY) is involved in neuroprotection as well as restoration of bone marrow dysfunction from chemotherapy induced nerve injury. However, the role of NPY in chemotherapy- induced nephrotoxicity has not been studied. Here, we show that NPY rescues renal dysfunction by reducing the expression of pro-apoptotic proteins in cisplatin induced nephrotoxicity through Y1 receptor, suggesting that NPY can protect kidney against cisplatin nephrotoxicity as a possible useful agent to prevent and treat cisplatin-induced nephrotoxicity. [BMB Reports 2016; 49(5): 288-292].


Assuntos
Apoptose/efeitos dos fármacos , Cisplatino/efeitos adversos , Nefropatias/tratamento farmacológico , Rim/patologia , Neuropeptídeo Y/uso terapêutico , Substâncias Protetoras/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Animais , Regulação para Baixo/efeitos dos fármacos , Feminino , Rim/efeitos dos fármacos , Nefropatias/induzido quimicamente , Nefropatias/genética , Masculino , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/farmacologia , Substâncias Protetoras/farmacologia , Receptores de Neuropeptídeo Y/metabolismo
7.
Brain Res ; 1646: 514-521, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27369450

RESUMO

With the increasing worldwide incidence of Alzheimer's disease (AD), there is a critical need for the discovery of more effective diagnostic methods. However, development of diagnostic tools in AD has been hindered by obstacles such as the absence of exact biomarkers. Apoptosis caused by amyloid-ß (Aß) plays an important role in AD pathology; therefore, provides an attractive biological target for the diagnosis of AD. The present study aimed to evaluate the potential of small peptide, named ApoPep-1 (Apoptosis-targeting peptide-1) as a new apoptosis imaging agent in AD. The fluorescein-conjugated ApoPep-1, but not the control peptide, targeted apoptotic cells in the brain of amyloid precursor protein (APP)/presenilin 1 (PS1) mice. We also observed fluorescence signals during in vivo imaging of apoptotic cells using ApoPep-1, and fluorescence levels increased in an age-dependent manner in APP/PS1 mice. Ex vivo imaging of isolated brains in APP/PS1 mice further confirmed the targeting of ApoPep-1 to apoptotic cells. The fluorescein-labeled ApoPep-1 co-localized with brain cells such as neurons, astrocytes, and microglia, all of which undergo apoptosis in the APP/PS1 mice brain. These findings demonstrate that ApoPep-1 can target apoptotic brain cells, and be used for experimental investigations relevant to apoptosis in AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/administração & dosagem , Apoptose , Encéfalo/fisiopatologia , Imuno-Histoquímica/métodos , Oligopeptídeos/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Doença de Alzheimer/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/fisiologia , Encéfalo/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Fluoresceína/administração & dosagem , Corantes Fluorescentes/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Microglia/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Oligopeptídeos/metabolismo
8.
BMB Rep ; 47(8): 439-44, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24314140

RESUMO

Inhibition of an increase of osteoclasts has become the most important treatment for osteoporosis. The CXCR4 antagonist, AMD3100, plays an important role in the mobilization of osteoclast precursors within bone marrow (BM). However, the actual therapeutic impact of AMD3100 in osteoporosis has not yet been ascertained. Here we demonstrate the therapeutic effect of AMD3100 in the treatment of ovariectomy-induced osteoporosis in mice. We found that treatment with AMD3100 resulted in direct induction of release of SDF-1 from BM to blood and mobilization of hematopoietic stem/progenitor cells (HSPCs) in an osteoporosis model. AMD3100 prevented bone density loss after ovariectomy by mobilization of HSPCs, suggesting a therapeutic strategy to reduce the number of osteoclasts on bone surfaces. These findings support the hypothesis that treatment with AMD3100 can result in efficient mobilization of HSPCs into blood through direct blockade of the SDF-1/CXCR4 interaction in BM and can be considered as a potential new therapeutic intervention for osteoporosis.


Assuntos
Células-Tronco Hematopoéticas/citologia , Compostos Heterocíclicos/uso terapêutico , Osteoporose/terapia , Animais , Benzilaminas , Conservadores da Densidade Óssea/farmacologia , Conservadores da Densidade Óssea/uso terapêutico , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular , Quimiocina CXCL12/metabolismo , Ciclamos , Modelos Animais de Doenças , Feminino , Transplante de Células-Tronco Hematopoéticas , Compostos Heterocíclicos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Osteoporose/tratamento farmacológico , Ovariectomia , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA