Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Brain Res ; 1736: 146779, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32171704

RESUMO

Inflammation plays an important role in acute and chronic cerebral ischemia. Recent reports indicate that the inflammatory response triggered by tissue damage is mediated by a multiple-protein complex called the inflammasome. The NOD-like receptor family, pyrin domain containing 3 (NLRP3) and absent in melanoma 2 (AIM2) inflammasome complex triggers caspase 1-mediated maturation of interleukin (IL)-1ß and IL-18. This study tested the hypothesis that chronic cerebral hypoperfusion activates inflammasomes in the white matter of the brain. To induce cerebral hypoperfusion, C57BL/6J mice were subjected to a sham or bilateral common carotid artery stenosis (BCAS) operation using microcoils with an internal diameter of 0.18 mm. At 2 and 4 weeks after BCAS, the mice were sacrificed (n = 5 in each group). Coronal sections were stained with anti-NLRP3 and anti-AIM2 antibodies. Activation of the inflammasome and cytokines was assessed using immunohistochemistry and cell counting. IL-18 and IL-1ß levels were determined by ELISA. Cell counting revealed an increase in NLRP3 and AIM2 inflammasomes at 2 and 4 weeks after BCAS. Immunoreactivity was observed in glial cells in the white matter and corpus callosum. IL-18 and IL-1ß concentrations were significantly increased compared with those in the sham operation group. Expression of NLRP3 and AIM2 was upregulated in glial cells in the autopsied brains of patients with cerebral infarction in the chronic phase. These results suggest that chronic cerebral hypoperfusion induces upregulation of NLRP3 and AIM2 inflammasomes; therefore, inflammasomes may play an important role in the sterile inflammatory response in astrocytes and microglia during chronic cerebral hypoperfusion.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Caspase 1/metabolismo , Proteínas de Ligação a DNA/fisiologia , Inflamassomos/metabolismo , Inflamação , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Perfusão/métodos , Traumatismo por Reperfusão/fisiopatologia , Substância Branca/metabolismo
2.
Sci Rep ; 10(1): 2341, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-32047216

RESUMO

Incretins, including glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), secreted from enteroendocrine cells after food ingestion, are currently recognized to regulate glucose metabolism through insulin secretion. We previously demonstrated that platelet-derived growth factor-BB (PDGF-BB) induces the migration of osteoblast-like MC3T3-E1 cells through mitogen-activated protein (MAP) kinases, including p38 MAP kinase. In the present study, we investigated whether or not incretins affect the osteoblast migration. The PDGF-BB-induced cell migration was significantly reinforced by GLP-1, GIP or cAMP analogues in MC3T3-E1 cells and normal human osteoblasts. The upregulated migration by GLP-1 or cAMP analogues was suppressed by H89, an inhibitor of protein kinase A. The amplification by GLP-1 of migration induced by PDGF-BB was almost completely reduced by SB203580, a p38 MAP kinase inhibitor in MC3T3-E1 cells and normal human osteoblasts. In addition, GIP markedly strengthened the PDGF-BB-induced phosphorylation of p38 MAP kinase. Exendin-4, a GLP-1 analogue, induced Rho A expression and its translocation from cytoplasm to plasma membranes in osteoblasts at the epiphyseal lines of developing mouse femurs in vivo. These results strongly suggest that incretins accelerates the PDGF-BB-induced migration of osteoblasts via protein kinase A, and the up-regulation of p38 MAP kinase is involved in this acceleration. Our findings may highlight the novel potential of incretins to bone physiology and therapeutic strategy against bone repair.


Assuntos
Becaplermina/metabolismo , Movimento Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Incretinas/farmacologia , Osteoblastos/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Becaplermina/genética , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/genética , Camundongos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Fosforilação , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/genética
3.
Neurosci Lett ; 704: 133-140, 2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-30954605

RESUMO

Alzheimer's disease (AD) is the most common type of dementia in aging adults. Increasing evidence has revealed that vascular risk factors influence the midlife development of AD and that diet-induced obesity accelerates tau phosphorylation in tau transgenic mice and increases the level of serum leptin receptor (leptin-R). Leptin-R is upregulated in the peri-infarct cortices after acute cerebral ischemia. Leptin may be protective against the development of AD as it can inactivate GSK-3ß through the phosphorylation of Ser-9, leading to the reduction of tau phosphorylation. Using tau transgenic mice, the present study examined whether chronic cerebral hypoperfusion affects leptin-R signaling and tau phosphorylation. Eight-month-old tau transgenic mice (T44) overexpressing the shortest human tau isoform were subjected to chronic cerebral hypoperfusion with bilateral common carotid artery stenosis (BCAS) using microcoils or sham surgery. Their brains were analyzed four weeks later to evaluate the expression of phosphorylated tau and leptin-R via immunohistochemistry and Western blot analysis. In addition, expression of leptin-R was examined in the rat primary astrocyte cultures subjected to prolonged chemical hypoxic stress, as well as in autopsied brains. BCAS upregulated leptin-R expression and promoted the expression of phosphorylated tau in T44 Tg mice. In primary astrocyte cultures, leptin-R was upregulated under hypoxic conditions via the phosphorylated AKT/pAKT pathway, possibly suppressing the expression of caspase 3. Leptin-R was also strongly expressed in autopsied brains with AD and cerebrovascular diseases. These results collectively indicate that chronic cerebral hypoperfusion promotes leptin-R signaling and tau phosphorylation.


Assuntos
Astrócitos/metabolismo , Isquemia Encefálica/metabolismo , Córtex Cerebral/metabolismo , Receptores para Leptina/metabolismo , Proteínas tau/metabolismo , Animais , Isquemia Encefálica/etiologia , Estenose das Carótidas/complicações , Córtex Cerebral/irrigação sanguínea , Masculino , Camundongos Transgênicos , Fosforilação , Regulação para Cima
4.
Stroke ; 45(12): 3698-703, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25370583

RESUMO

BACKGROUND AND PURPOSE: Proper brain function is maintained by an integrated system called the neurovascular unit (NVU) comprised cellular and acellular elements. Although the individual features of specific neurovascular components are understood, it is unknown how they respond to ischemic stress as a functional unit. Therefore, we established an in vivo imaging method and clarified the NVU response to chronic cerebral hypoperfusion. METHODS: Green mice (b-act-EGFP) with SR101 plasma labeling were used in this experiment. A closed cranial window was made over the left somatosensory cortex. To mimic chronic cerebral hypoperfusion, mice were subjected to bilateral common carotid artery stenosis operations using microcoils. In vivo real-time imaging was performed using 2-photon laser-scanning microscopy during the preoperative period, and after 1 day and 1 and 2 weeks of bilateral common carotid artery stenosis or sham operations. RESULTS: Our method allowed 3-dimensional observation of most of the components of the NVU, as well as dynamic capillary microcirculation. Under chronic cerebral hypoperfusion, we did not detect any structural changes of each cellular component in the NVU; however, impairment of microcirculation was detected over a prolonged period. In the pial small arteries and veins, rolling and adhesion of leukocyte were detected, more prominently in the latter. In the deep cortical capillaries, flow stagnation because of leukocyte plugging was frequently observed. CONCLUSIONS: We established an in vivo imaging method for real-time visualization of the NVU. It seems that under chronic cerebral hypoperfusion, leukocyte activation has a critical role in microcirculation disturbance.


Assuntos
Isquemia Encefálica/patologia , Encéfalo/patologia , Neuroimagem/métodos , Animais , Proteínas de Fluorescência Verde/genética , Imageamento Tridimensional/métodos , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Confocal/métodos
5.
PLoS One ; 9(6): e100045, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24936646

RESUMO

Inflammation is crucially involved in the development of carotid plaques. We examined the relationship between plaque vulnerability and inflammatory biomarkers using intraoperative blood and tissue specimens. We examined 58 patients with carotid stenosis. Following carotid plaque magnetic resonance imaging, 41 patients underwent carotid artery stenting (CAS) and 17 underwent carotid endarterectomy (CEA). Blood samples were obtained from the femoral artery (systemic) and common carotid artery immediately before and after CAS (local). Seventeen resected CEA tissue samples were embedded in paraffin, and histopathological and immunohistochemical analyses for IL-6, IL-10, E-selectin, adiponectin, and pentraxin 3 (PTX3) were performed. Serum levels of IL-6, IL-1ß, IL-10, TNFα, E-selectin, VCAM-1, adiponectin, hs-CRP, and PTX3 were measured by multiplex bead array system and ELISA. CAS-treated patients were classified as stable plaques (n = 21) and vulnerable plaques (n = 20). The vulnerable group showed upregulation of the proinflammatory cytokines (IL-6 and TNFα), endothelial activation markers (E-selectin and VCAM-1), and inflammation markers (hs-CRP and PTX3) and downregulation of the anti-inflammatory markers (adiponectin and IL-10). PTX3 levels in both systemic and intracarotid samples before and after CAS were higher in the vulnerable group than in the stable group. Immunohistochemical analysis demonstrated that IL-6 was localized to inflammatory cells in the vulnerable plaques, and PTX3 was observed in the endothelial and perivascular cells. Our findings reveal that carotid plaque vulnerability is modulated by the upregulation and downregulation of proinflammatory and anti-inflammatory factors, respectively. PTX3 may thus be a potential predictive marker of plaque vulnerability.


Assuntos
Proteína C-Reativa/análise , Estenose das Carótidas/diagnóstico , Mediadores da Inflamação/análise , Placa Aterosclerótica/diagnóstico , Componente Amiloide P Sérico/análise , Idoso , Biomarcadores/análise , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade
6.
Free Radic Res ; 48(6): 694-705, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24697733

RESUMO

In Parkinson's disease (PD), oxidative stresses cause cell death of dopaminergic neurons of the substantia nigra (SN), but its molecular mechanism still remains unclarified. Our previous study of proteomic analysis in the monkey CA1 hippocampus after ischemia-reperfusion revealed reactive oxygen species (ROS)-induced carbonyl modification of a molecular chaperone, heat shock 70-kDa protein 1 (Hsp70.1), especially in its key site, Arg469. Here, to clarify the mechanism of neurodegeneration in PD, the SN tissue of the same monkey experimental paradigm was studied for identifying and characterizing carbonylated proteins by the two-dimensional gel electrophoresis with immunochemical detection of protein carbonyls (2D Oxyblot). We found carbonyl modification not only of Hsp70.1 but also of mitochondrial aconitase, dihydropyrimidinase-related protein 2, T-complex protein 1 subunit alpha, dihydrolipoyl dehydrogenase, fructose-bisphosphate aldolase C, glutamate dehydrogenase 1, and aspartate aminotransferase. Intriguingly, in the SN also, the carbonylation site of Hsp70.1 was identified to be Arg469. Since Hsp70.1 is recently known to stabilize the lysosomal membrane, its oxidative injury conceivably plays an important role in the ROS-mediated neuronal cell death by inducing lysosomal destabilization. Implications of each carbonylated proteins for the dopaminergic neuronal death were discussed, in comparison with the CA1 neuronal death.


Assuntos
Neurônios Dopaminérgicos/patologia , Mitocôndrias/metabolismo , Carbonilação Proteica , Traumatismo por Reperfusão/patologia , Substância Negra/patologia , Animais , Apoptose , Proteínas de Choque Térmico HSP70/metabolismo , Lisossomos/patologia , Macaca , Mitocôndrias/enzimologia , Estresse Oxidativo , Doença de Parkinson/patologia , Proteômica , Espécies Reativas de Oxigênio/metabolismo , Substância Negra/citologia
7.
Brain Res ; 1533: 131-40, 2013 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-23954745

RESUMO

Transactivation-responsive DNA-binding protein 43 (TDP-43) is closely involved in the pathogenesis of frontotemporal lobar degeneration. The native form, but not phosphorylated form, of TDP-43 has been reported to redistribute during acute neuronal injuries. Here, we examined whether the expression of phosphorylated TDP-43 was altered following chronic neuronal injury. C57BL/6 mice were subjected to sham operation or bilateral common carotid artery stenosis (BCAS) using microcoils, and changes in proteolytic cleavage, phosphorylation, and subcellular redistribution of TDP-43 were examined by immunoblotting and immunohistochemistry. We also monitored the expression of importin ß, which is involved in the transport of TDP-43. Immunoblotting showed an increase in phosphorylated TDP-43 in the nuclear fraction after BCAS using microcoils. Moreover, immunoreactivity toward phosphorylated TDP-43 was observed in the neuronal cytoplasm in the cerebral cortex and hippocampus, and importin ß levels decreased after the operation. Immunoreactivity toward phosphorylated TDP-43 was partly colocalized with immunoreactivity toward caspase 3 in the neuronal cytoplasm. These results suggested that chronic cerebral ischemia induced redistribution and abnormal phosphorylation of TDP-43, which may be triggered by downregulation of importin ß and may partly result in neuronal death.


Assuntos
Isquemia Encefálica/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neurônios/metabolismo , Animais , Apoptose , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/patologia , Doença Crônica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Fosforilação , beta Carioferinas/metabolismo
8.
Eur Neurol ; 70(1-2): 70-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23796701

RESUMO

Pigmented neurons in the substantia nigra pars compacta (SNc) and locus coeruleus (LC) show decreased numbers differentially in Parkinson's disease (PD) and multiple system atrophy (MSA). Recent reports have described that fast spin-echo T1-weighted magnetic resonance imaging (MRI) by a 3-tesla machine can visualize neuromelanin-related contrast of the noradrenergic and dopaminergic neurons respectively in the LC and the SNc. Using neuromelanin MRI at 3 T, we investigated possible alterations of these catecholaminergic neurons in 32 PD and 9 MSA patients, and compared the results with those of 23 normal volunteers. The contrast ratio of the LC and SNc was decreased in MSA and PD patients, most prominently in the LC in MSA patients. The contrast ratio of the SNc was correlated with the Hoehn-Yahr stage of PD and the severity of neuroradiological abnormalities in MSA. These results indicate a potential diagnostic value of neuromelanin MRI to distinguish MSA patients from normal and PD patients.


Assuntos
Encéfalo/patologia , Imageamento por Ressonância Magnética/métodos , Melaninas , Atrofia de Múltiplos Sistemas/diagnóstico , Doença de Parkinson/diagnóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Melaninas/análise , Pessoa de Meia-Idade
9.
ACS Chem Neurosci ; 4(8): 1183-93, 2013 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-23668665

RESUMO

Disruption of the blood-brain barrier (BBB) can occur in various pathophysiological conditions. Administration of extraneous tracers that can pass the disrupted, but not the intact, BBB and detection of the extravasation have been widely used to assess BBB disruption in animal models. Although several fluorescent tracers have been successfully used, the administration of these tracers basically requires intravascular injection, which can be laborious when using small animals such as zebrafish. To identify fluorescent tracers that could be easily administered into various animal models and visualize the BBB disruption in vivo, we prepared nine structurally related indoline derivatives (IDs) as a minimum set of diverse fluorescent compounds. We found that one ID, ZMB741, had the highest affinity for serum albumin and emitted the strongest fluorescence in the presence of serum albumin of the nine IDs tested. The affinity to serum albumin and the fluorescence intensity was superior to those of Evans blue and indocyanine green that have been conventionally used to assess the BBB disruption. We showed that ZMB741 could be administered into zebrafish by static immersion or mice by intraperitoneal injection and visualizes the active disruption of their BBB. These results suggest that ZMB741 can be a convenient and versatile tool for in vivo fluorescent imaging of BBB disruption in various animal models. The strategy used in this study can also be applied to diversity-oriented libraries to identify novel fluorescent tracers that may be superior to ZMB741.


Assuntos
Barreira Hematoencefálica/metabolismo , Diagnóstico por Imagem/métodos , Corantes Fluorescentes/farmacocinética , Indóis/farmacocinética , Animais , Transporte Biológico , Barreira Hematoencefálica/efeitos dos fármacos , Modelos Animais de Doenças , Corantes Fluorescentes/administração & dosagem , Indóis/administração & dosagem , Camundongos , Permeabilidade , Albumina Sérica/química , Peixe-Zebra
10.
Neuropathology ; 33(2): 199-203, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22882713

RESUMO

Over 100 mutations have been described in the presenilin-1 gene (PSEN1), resulting in familial Alzheimer disease (AD). However, of the limited number of autopsy cases, only one has been reported from an AD family with an L420R PSEN1 mutation. We describe here clinical and neuropathological features of a patient with dementia-parkinsonism from a family with a PSEN1 mutation (L420R). A 43-year-old Japanese woman was autopsied 12 years after the onset of her progressive dementia and 4 years after the onset of parkinsonism. Throughout the neocortex and hippocampus, cotton wool plaques were identified, densely packed, in almost all the cortical layers along with neuronal loss, gliosis, NFT and neuropil threads. In addition, CAA affecting meningeal, subpial and cortical arterioles was found, as well as amyloid ß-protein (Aß)-deposition in the capillaries (capillary CAA) in the neocortex and subcortical nuclei. There was loss of pigmented neurons in the substantia nigra. The putamen was densely packed with diffuse plaques and rarely showed capillary CAA, whereas the globus pallidus showed extensive capillary CAA but no plaques. This differential distribution is similar to that reported for a previous patient with a mutation in PSEN1. It is concluded that neuropathological changes in the substantia nigra and lenticular nuclei were responsible for the patient's parkinsonism. Capillary transport of Aß unique to the respective tissue of the patient may result in the differential distribution of Aß between the putamen and globus pallidus seen in individuals with a PSEN1 mutation.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Mutação/genética , Presenilina-1/genética , Encéfalo/patologia , Evolução Fatal , Feminino , Humanos , Pessoa de Meia-Idade
11.
PLoS One ; 7(2): e31594, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22389670

RESUMO

BACKGROUND: TNF-related apoptosis-inducing ligand/Apo2 ligand (TRAIL/Apo2L) selectively induces apoptosis in various cancer cells including myeloma (MM) cells. However, the susceptibility of MM cells to TRAIL is largely low in most of MM cells by yet largely unknown mechanisms. Because TNF-α converting enzyme (TACE) can cleave some TNF receptor family members, in the present study we explored the roles of proteolytic modulation by TACE in TRAIL receptor expression and TRAIL-mediated cytotoxicity in MM cells. METHODOLOGY/PRINCIPAL FINDINGS: MM cells preferentially expressed death receptor 4 (DR4) but not DR5 on their surface along with TACE. Conditioned media from RPMI8226 and U266 cells contained a soluble form of DR4. The DR4 levels in these conditioned media were reduced by TACE inhibition by the TACE inhibitor TAPI-0 as well as TACE siRNA. Conversely, the TACE inhibition restored surface levels of DR4 but not DR5 in these cells without affecting DR4 mRNA levels. The TACE inhibition was able to restore cell surface DR4 expression in MM cells even in the presence of bone marrow stromal cells or osteoclasts, and enhanced the cytotoxic effects of recombinant TRAIL and an agonistic antibody against DR4 on MM cells. CONCLUSIONS/SIGNIFICANCE: These results demonstrate that MM cells post-translationally down-modulate the cell surface expression of DR4 through ectodomain shedding by endogenous TACE, and that TACE inhibition is able to restore cell surface DR4 levels and the susceptibility of MM cells to TRAIL or an agonistic antibody against DR4. Thus, TACE may protect MM cells from TRAIL-mediated death through down-modulation of cell-surface DR4. It can be envisaged that TACE inhibition augments clinical efficacy of TRAIL-based immunotherapy against MM, which eventually becomes resistant to the present therapeutic modalities.


Assuntos
Proteínas ADAM/metabolismo , Mieloma Múltiplo/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/genética , Proteína ADAM17 , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Citometria de Fluxo , Células HL-60 , Humanos , Mieloma Múltiplo/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células U937
12.
Br J Haematol ; 155(3): 328-39, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21902681

RESUMO

The spicamycin analogue KRN5500 alters glycoprotein processing and induces damage in the endoplasmic reticulum (ER)-Golgi apparatus in cancer cells. In the present study, we explored the cytotoxic effects of KRN5500 on multiple myeloma (MM) cells and the bone marrow microenvironment with special reference to ER stress. Cell proliferation assay showed that KRN5500 induced G1 arrest and apoptosis in MM cells in a time- and dose-dependent manner. KRN5500 enhanced ER stress independently of caspase activation in MM cells. This cell death was observed even in the presence of bone marrow stroma cells or osteoclasts. Notably, KRN5500 induced cell death also in osteoclasts. In vivo effects of KRN5500 were evaluated using two xenograft models established in severe combined immunodeficient (SCID) mice by either subcutaneous injection of RPMI 8226 cells or intra-bone injection of INA-6 cells to subcutaneously implanted rabbit bones (SCID-rab model). KRN5500 significantly inhibited tumour growth in both animal models, and decreased the number of osteoclasts, which resulted in prevention of bone destruction in the SCID-rab model. These results suggest that KRN5500 exerts anti-MM effects through impairing both MM cells and osteoclasts. Therefore, this unique mechanism of KRN5500 might be a useful therapeutic option in patients with MM.


Assuntos
Mieloma Múltiplo/tratamento farmacológico , Osteoclastos/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/patologia , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Fase G1/efeitos dos fármacos , Humanos , Camundongos , Camundongos SCID , Mieloma Múltiplo/patologia , Osteoclastos/patologia , Nucleosídeos de Purina/farmacologia , Coelhos , Distribuição Aleatória , Células Estromais/patologia , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Hypertension ; 58(4): 635-42, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21859961

RESUMO

The role of the renin-angiotensin system in cognitive impairment is unclear. This work was undertaken to test our hypothesis that renin-angiotensin system may contribute to cognitive decline and brain damage caused by chronic cerebral ischemia. C57BL/6J mice were subjected to bilateral common carotid artery stenosis with microcoil to prepare mice with chronic cerebral hypoperfusion, a model of subcortical vascular dementia. The effects of aliskiren, a direct renin inhibitor, or Tempol, a superoxide scavenger, on brain damage and working memory in these mice were examined. Chronic cerebral hypoperfusion significantly increased brain renin activity and angiotensinogen expression in C57BL/6J mice, which was attributed to the increased renin in activated astrocytes and microvessels and the increased angiotensinogen in activated astrocytes in white matter. Aliskiren pretreatment significantly inhibited brain renin activity and ameliorated brain p67(phox)-related NADPH oxidase activity, oxidative stress, glial activation, white matter lesion, and spatial working memory deficits in C57BL/6J mice with bilateral common carotid artery stenosis. To elucidate the role of oxidative stress in brain protective effects of aliskiren, we also examined the effect of Tempol in the same mice with bilateral common carotid artery stenosis. Tempol pretreatment mimicked the brain protective effects of aliskiren in this mouse model. Posttreatment of mice with aliskiren or Tempol after bilateral common carotid artery stenosis also prevented cognitive decline. In conclusion, chronic cerebral hypoperfusion induced the activation of the brain renin-angiotensin system. Aliskiren ameliorated brain damage and working memory deficits in the model of chronic cerebral ischemia through the attenuation of oxidative stress. Thus, direct renin inhibition seems to be a promising therapeutic strategy for subcortical vascular dementia.


Assuntos
Dano Encefálico Crônico/prevenção & controle , Isquemia Encefálica/metabolismo , Transtornos Cognitivos/prevenção & controle , Demência Vascular/metabolismo , Renina/antagonistas & inibidores , Renina/metabolismo , Amidas/farmacologia , Angiotensinogênio/metabolismo , Animais , Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Dano Encefálico Crônico/epidemiologia , Dano Encefálico Crônico/metabolismo , Transtornos Cognitivos/epidemiologia , Transtornos Cognitivos/metabolismo , Óxidos N-Cíclicos/farmacologia , Modelos Animais de Doenças , Fumaratos/farmacologia , Hipertensão/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , Fatores de Risco , Marcadores de Spin
14.
Brain Res ; 1410: 12-23, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21803337

RESUMO

Hyperphosphorylated tau protein constitutes a significant portion of intracellular inclusions in some neurodegenerative diseases. In addition, mutations in tau protein cause familial forms of frontotemporal dementia (FTD), indicating that dysfunction of tau protein is responsible for neurodegeneration and dementia. P301S tau-transgenic (Tg) mouse expressing human mutant tau in neurons exhibits similar features of human tauopathies including neuronal degeneration and filament accumulation consisted of hyperphosphorylated tau protein. In the present study, we attempted to characterize protein expression profiles in P301S tau-Tg mouse by using two-dimensional differential in-gel electrophoresis (2D-DIGE) coupled by peptide mass fingerprinting (PMF). As a result, we identified four upregulated proteins; heat shock protein 27 (Hsp27), peroxiredoxin 6 (Prdx6), apolipoprotein E (ApoE), and latexin (LTXN), all of which may function as a neuroprotective mechanism against tau toxicity. In immunohistochemistry, these four proteins were increased invariably in astrocytes, and these astrocytes infiltrated the area in which there are numerous accumulations of hyperphosphorylated tau and neuronal loss. Therefore, these results may indicate that astrocytes provide a neuroprotective mechanism against tau toxicity.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Citoproteção/fisiologia , Degeneração Neural/metabolismo , Neurônios/metabolismo , Proteínas tau/metabolismo , Animais , Antígenos/metabolismo , Apolipoproteínas E/metabolismo , Astrócitos/patologia , Encéfalo/patologia , Morte Celular , Modelos Animais de Doenças , Proteínas de Choque Térmico HSP27/metabolismo , Camundongos , Camundongos Transgênicos , Degeneração Neural/genética , Degeneração Neural/patologia , Neurônios/patologia , Peroxirredoxina VI/metabolismo , Fosforilação , Tauopatias/genética , Tauopatias/metabolismo , Regulação para Cima , Proteínas tau/genética
15.
Int J Hematol ; 94(1): 63-70, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21698356

RESUMO

Multiple myeloma (MM) cells stimulate osteoclastogenesis, and osteoclasts (OCs) in turn enhance MM growth and drug resistance, resulting in a vicious cycle. Vγ9Vδ2 T cells exert potent anti-tumor effects, making T cell-based immunotherapies using these cells attractive candidates for currently incurable malignancies, such as MM. However, the impact of such treatments on the MM-OC interaction is largely unknown. We demonstrate here that Vγ9Vδ2 T cells expanded by zoledronic acid and IL-2 exerted potent cytotoxic effects on both MM cells and OCs, even in coculture settings, but showed no such effect on bone marrow stromal cells. Vγ9Vδ2 T cells marginally affected colony formation from normal hematopoietic progenitors, and furthermore migrated toward osteopontin and MIP-1α, factors produced by the MM-OC interaction. These results suggest that Vγ9Vδ2 T cells expanded by zoledronic acid and IL-2 are able to migrate to MM bone lesions and preferentially target OCs as well as MM cells, thereby inhibiting both tumor expansion and bone destruction.


Assuntos
Comunicação Celular/imunologia , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Osteoclastos/efeitos dos fármacos , Linfócitos T/imunologia , Comunicação Celular/efeitos dos fármacos , Técnicas de Cultura de Células/métodos , Movimento Celular/efeitos dos fármacos , Citotoxicidade Imunológica , Difosfonatos/farmacologia , Humanos , Imidazóis/farmacologia , Interleucina-2/farmacologia , Mieloma Múltiplo/imunologia , Osteoclastos/patologia , Receptores de Antígenos de Linfócitos T gama-delta , Linfócitos T/patologia , Ácido Zoledrônico
16.
Rinsho Ketsueki ; 51(8): 690-5, 2010 Aug.
Artigo em Japonês | MEDLINE | ID: mdl-20805677

RESUMO

A 67-year-old man was diagnosed with multiple myeloma IgA-lambda type, Durie-Salmon classification stage IIIA in October 2001. He received five courses of induction chemotherapy consisting of vincristine, doxorubicin and dexamethasone and then underwent high dose chemotherapy followed by autologous stem cell transplantation in March 2003. He achieved partial response, but then relapsed after treatment with thalidomide and was admitted to our hospital in June 2007. The patient was complicated by tumor lysis syndrome (TLS) after receiving bortezomib therapy twice. Computed tomography after bortezomib therapy showed the rapid appearance of tumors in the right upper lobe of the lung, tail of the pancreas and the spleen. Though he was treated with antifungal agents, micafungin and voriconazole, he died eighty-five days after admission. Autopsy specimen showed fungal clumps and hemorrhagic infarction in the lung and spleen, and vegetation at the mitral valve was the same fungus as found in the lung. We diagnosed disseminated zygomycosis based on the pathological fungal morphology. This case suggested that metabolic acidosis was caused by TLS, while poorly controlled diabetes, secondary hemochromatosis due to transfusion, and breakthrough zygomycosis during antifungal therapy were thought to be factors contributing to the development of zygomycosis.


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Ácidos Borônicos/efeitos adversos , Ácidos Borônicos/uso terapêutico , Mieloma Múltiplo/complicações , Mieloma Múltiplo/tratamento farmacológico , Pirazinas/efeitos adversos , Pirazinas/uso terapêutico , Zigomicose/etiologia , Zigomicose/patologia , Acidose/etiologia , Idoso , Antifúngicos/efeitos adversos , Antifúngicos/uso terapêutico , Bortezomib , Equinocandinas/efeitos adversos , Equinocandinas/uso terapêutico , Evolução Fatal , Humanos , Lipopeptídeos/efeitos adversos , Lipopeptídeos/uso terapêutico , Masculino , Micafungina , Pirimidinas/efeitos adversos , Pirimidinas/uso terapêutico , Triazóis/efeitos adversos , Triazóis/uso terapêutico , Síndrome de Lise Tumoral/etiologia , Voriconazol , Zigomicose/tratamento farmacológico
17.
PLoS One ; 5(3): e9870, 2010 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-20360846

RESUMO

BACKGROUND: Multiple myeloma (MM) expands almost exclusively in the bone marrow and generates devastating bone lesions, in which bone formation is impaired and osteoclastic bone resorption is enhanced. TGF-beta, a potent inhibitor of terminal osteoblast (OB) differentiation, is abundantly deposited in the bone matrix, and released and activated by the enhanced bone resorption in MM. The present study was therefore undertaken to clarify the role of TGF-beta and its inhibition in bone formation and tumor growth in MM. METHODOLOGY/PRINCIPAL FINDINGS: TGF-beta suppressed OB differentiation from bone marrow stromal cells and MC3T3-E1 preosteoblastic cells, and also inhibited adipogenesis from C3H10T1/2 immature mesenchymal cells, suggesting differentiation arrest by TGF-beta. Inhibitors for a TGF-beta type I receptor kinase, SB431542 and Ki26894, potently enhanced OB differentiation from bone marrow stromal cells as well as MC3T3-E1 cells. The TGF-beta inhibition was able to restore OB differentiation suppressed by MM cell conditioned medium as well as bone marrow plasma from MM patients. Interestingly, TGF-beta inhibition expedited OB differentiation in parallel with suppression of MM cell growth. The anti-MM activity was elaborated exclusively by terminally differentiated OBs, which potentiated the cytotoxic effects of melphalan and dexamethasone on MM cells. Furthermore, TGF-beta inhibition was able to suppress MM cell growth within the bone marrow while preventing bone destruction in MM-bearing animal models. CONCLUSIONS/SIGNIFICANCE: The present study demonstrates that TGF-beta inhibition releases stromal cells from their differentiation arrest by MM and facilitates the formation of terminally differentiated OBs, and that terminally differentiated OBs inhibit MM cell growth and survival and enhance the susceptibility of MM cells to anti-MM agents to overcome the drug resistance mediated by stromal cells. Therefore, TGF-beta appears to be an important therapeutic target in MM bone lesions.


Assuntos
Mieloma Múltiplo/patologia , Osteoblastos/citologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Células 3T3 , Animais , Apoptose , Células da Medula Óssea/citologia , Osso e Ossos/patologia , Diferenciação Celular , Proliferação de Células , Dexametasona/farmacologia , Masculino , Melfalan/farmacologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos SCID , Mieloma Múltiplo/metabolismo , Coelhos , Receptores de Interleucina-6/genética , Fator de Crescimento Transformador beta/metabolismo
18.
Blood ; 114(20): 4517-26, 2009 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19762488

RESUMO

Monocytes give rise to macrophages, osteoclasts (OCs), and dendritic cells (DCs). Macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-kappaB (RANK) ligand induce OC differentiation from monocytes, whereas granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4) trigger monocytic differentiation into DCs. However, regulatory mechanisms for the polarization of monocytic differentiation are still unclear. The present study was undertaken to clarify the mechanism of triggering the deflection of OC and DC differentiation from monocytes. GM-CSF and IL-4 abolished monocytic differentiation into OCs while inducing DC differentiation even in the presence of M-CSF and RANK ligand. GM-CSF and IL-4 in combination potently up-regulate tumor necrosis factor-alpha (TNF-alpha) converting enzyme (TACE) and activity in monocytes, causing ectodomain shedding of M-CSF receptor, resulting in the disruption of its phosphorylation by M-CSF as well as the induction of osteoclastogenesis from monocytes by M-CSF and RANK ligand. Interestingly, TACE inhibition robustly causes the resumption of the surface expression of M-CSF receptor on monocytes, facilitating M-CSF-mediated phosphorylation of M-CSF receptor and macrophage/OC differentiation while impairing GM-CSF- and IL-4-mediated DC differentiation from monocytes. These results reveal a novel proteolytic regulation of M-CSF receptor expression in monocytes to control M-CSF signaling and monocytic differentiation into macrophage/OC-lineage cells or DCs.


Assuntos
Proteínas ADAM/metabolismo , Diferenciação Celular/fisiologia , Células Dendríticas/citologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interleucina-4/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Proteína ADAM17 , Western Blotting , Células Dendríticas/metabolismo , Imunofluorescência , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Monócitos/citologia , Monócitos/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
19.
Int J Hematol ; 89(2): 223-226, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19225725

RESUMO

We report a patient with refractory multiple myeloma (MM) who developed platelet transfusion refractoriness (PTR). A 61-year-old woman was diagnosed with MM in July 2003. She underwent high-dose chemotherapy followed by autologous stem cell transplantation, and achieved a very good partial response. However, she relapsed in June 2006, and was referred to our hospital in October of the same year. Laboratory examinations showed pancytopenia and increased plasma cells in the peripheral blood. Platelet transfusions from random donors became ineffective, and anti-HLA class I antibody (83.8% positive) was detected in the serum by flow cytometry assay (Flow PRA). Therefore, she was considered to have developed PTR due to anti-HLA class I antibody caused by the previous blood transfusions. She was transfused with HLA-matched platelets, and then treated with bortezomib plus dexamethasone (BD) for refractory MM. The serum IgG level decreased from 7,451 to 1,735 mg/dL, and HLA class I antibody was markedly decreased to 1.9%. In addition, platelet transfusion from random donors showed clinical effects after BD therapy. This case suggests that bortezomib might be effective in different types of immune disease by inhibiting allo-reactive antibody.


Assuntos
Ácidos Borônicos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Pirazinas/uso terapêutico , Anticorpos , Bortezomib , Dexametasona/uso terapêutico , Feminino , Antígenos HLA/imunologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Pessoa de Meia-Idade , Mieloma Múltiplo/cirurgia , Transfusão de Plaquetas , Terapia de Salvação , Transplante Homólogo
20.
Clin Calcium ; 18(4): 438-46, 2008 Apr.
Artigo em Japonês | MEDLINE | ID: mdl-18379024

RESUMO

Myeloma cells stimulate bone resorption by enhancing osteoclast formation and suppress bone formation by inhibiting osteoblast differentiation. Macrophage inflammatory protein (MIP)-1alpha and MIP-1beta as well as RANK ligand play a major role in the enhancement of bone resorption in myeloma. Myeloma cell-derived soluble Wnt inhibitors as well as TGF-beta released from the bone tissues through enhanced bone resorption are thought to suppress osteoblast differentiation. Such pathognomonically skewed cellular components in the bone marrow create a microenvironment suitable for myeloma cell growth and survival (a myeloma niche) , which should be targeted to suppress myeloma expansion along with amelioration of bone lesions.


Assuntos
Reabsorção Óssea/etiologia , Mieloma Múltiplo/complicações , Reabsorção Óssea/patologia , Osso e Ossos/patologia , Diferenciação Celular , Quimiocina CCL3/fisiologia , Quimiocina CCL4/fisiologia , Humanos , Mieloma Múltiplo/patologia , Osteoblastos/citologia , Osteoclastos/fisiologia , Osteogênese , Ligante RANK/fisiologia , Fator de Crescimento Transformador beta/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA