Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Audiol Neurootol ; 28(1): 43-51, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36075188

RESUMO

INTRODUCTION: Loss of hair cells and degeneration of spiral ganglion neurons (SGN) lead to severe hearing loss or deafness. The successful use of a cochlear implant (CI) depends among other factors on the number of surviving SGN. Postoperative formation of fibrous tissue around the electrode array causes an increase in electrical impedances at the stimulating contacts. The use of immunophilin inhibitors may reduce the inflammatory processes without suppressing the immune response. Here, we report on in vitro experiments with different concentrations of immunophilin inhibitors MM284 and compound V20 regarding a possible application of these substances in the inner ear. METHODS: Standard cell lines (NIH/3T3 fibroblasts), freshly isolated SGN, and fibroblasts from neonatal rat cochleae (p3-5) were incubated with different concentrations of immunophilin inhibitors for 48 h. Metabolic activity of fibroblasts was investigated by MTT assay and cell survival by counting of immunochemically stained neurons and compared to controls. RESULTS: MM284 did not affect SGN numbers and neurite growth at concentrations of 4 × 10-5 mol/L and below, whereas V20 had no effect at 8 × 10-6 mol/L and below. Metabolic activity of fibroblasts was unchanged at these concentrations. CONCLUSION: Especially MM284 might be considered as a possible candidate for application within the cochlea.


Assuntos
Implantes Cocleares , Gânglio Espiral da Cóclea , Ratos , Animais , Imunofilinas/farmacologia , Cóclea , Neurônios , Fibroblastos
2.
Theriogenology ; 192: 62-72, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-36063671

RESUMO

Oocyte in vitro maturation (IVM) and vitrification procedures lead to detrimental effects on the overall oocyte quality. The addition of antioxidants during IVM, such as the coenzyme Q10 (Q10), has been demonstrated to positively impact on the cumulus-oocyte complexes due to its role in protection from oxidative damage and modulating gene transcription. Furthermore, glucocorticoids (GC) regulate gene transcription, energy metabolism and apoptosis during the early steps of reproduction. In this sense, most GC actions are mediated by the glucocorticoid receptor (NR3C1), a transcription factor. However, the specific roles of GC in ovarian physiology and oocyte maturation are still unknown. In this regard, a better knowledge on the expression of GC-related and apoptosis-related genes during IVM and cryopreservation procedures could potentially benefit the refinement of assisted reproductive techniques in the bovine species. The present study aims to explore the expression of NR3C1 mRNA in fresh and vitrified bovine oocytes and cumulus cells in response to Q10 (50 µM), and the effect of cortisol addition (0.25 µM, 0.5 µM) on the expression of NR3C1. We also studied the mRNA expression of NR3C1-related genes belonging to the GC regulation pathway, such as hydroxysteroid dehydrogenases (HSD11B1; HSD11B2), immunophilins (FKBP4; FKBP5), signal transducers and activators of transcription (STAT3; STAT5A), the mineralocorticoid receptor (NR3C2), and to the apoptosis pathway, such as the anti- (BCL2) and pro-apoptotic (BAX) mRNA transcripts in oocytes and cumulus cells 1) after IVM, and 2) after vitrification, both in presence or absence of Q10 supplementation during IVM. Our results show that there is an increase in the NR3C1 receptor expression after vitrification of oocytes, but not after exogenous cortisol supplementation during IVM. In addition, Q10 reduces the mRNA expression of HSD11B1 and FKBP5 in oocytes at levels of immature oocytes (HSD11B1 mRNA expression also in cumulus cells), and the BAX:BCL2 ratio mRNA expression. After vitrification in the presence of Q10, HSD11B2 mRNA expression increases in cumulus cells, while HSD11B1 and BAX:BCL2 mRNA expression decreases significantly both in oocytes and cumulus cells. In conclusion, our results show for the first time the effect of IVM, vitrification and Q10 supplementation on the mRNA relative expression of GC-related and apoptosis genes, and the effect of vitrification in the protein expression of NR3C1.


Assuntos
Células do Cúmulo , Vitrificação , Animais , Apoptose , Bovinos , Células do Cúmulo/fisiologia , Suplementos Nutricionais , Feminino , Glucocorticoides/metabolismo , Glucocorticoides/farmacologia , Hidrocortisona/metabolismo , Hidroxiesteroide Desidrogenases/metabolismo , Hidroxiesteroide Desidrogenases/farmacologia , Imunofilinas/metabolismo , Imunofilinas/farmacologia , Técnicas de Maturação in Vitro de Oócitos/métodos , Técnicas de Maturação in Vitro de Oócitos/veterinária , Oócitos/fisiologia , RNA Mensageiro/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Fatores de Transcrição/metabolismo , Ubiquinona/análogos & derivados , Proteína X Associada a bcl-2/metabolismo
3.
BMC Cancer ; 19(1): 351, 2019 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-30975104

RESUMO

BACKGROUND: Optimising breast cancer treatment remains a challenge. Resistance to therapy is a major problem in both ER- and ER+ breast cancer. Tumour recurrence after chemotherapy and/or targeted therapy leads to more aggressive tumours with enhanced metastatic ability. Self-renewing cancer stem cells (CSCs) have been implicated in treatment resistance, recurrence and the development of metastatic disease. METHODS: In this study, we utilised in vitro, in vivo and ex vivo breast cancer models using ER+ MCF-7 and ER- MDA-MB-231 cells, as well as solid and metastatic breast cancer patient samples, to interrogate the effects of FKBPL and its peptide therapeutics on metastasis, endocrine therapy resistant CSCs and DLL4 and Notch4 expression. The effects of FKBPL overexpression or peptide treatment were assessed using a t-test or one-way ANOVA with Dunnett's multiple comparison test. RESULTS: We demonstrated that FKBPL overexpression or treatment with FKBPL-based therapeutics (AD-01, pre-clinical peptide /ALM201, clinical peptide) inhibit i) CSCs in both ER+ and ER- breast cancer, ii) cancer metastasis in a triple negative breast cancer metastasis model and iii) endocrine therapy resistant CSCs in ER+ breast cancer, via modulation of the DLL4 and Notch4 protein and/or mRNA expression. AD-01 was effective at reducing triple negative MDA-MB-231 breast cancer cell migration (n ≥ 3, p < 0.05) and invasion (n ≥ 3, p < 0.001) and this was translated in vivo where AD-01 inhibited breast cancer metastasis in MDA-MB-231-lucD3H1 in vivo model (p < 0.05). In ER+ MCF-7 cells and primary breast tumour samples, we demonstrated that ALM201 inhibits endocrine therapy resistant mammospheres, representative of CSC content (n ≥ 3, p < 0.05). Whilst an in vivo limiting dilution assay, using SCID mice, demonstrated that ALM201 alone or in combination with tamoxifen was very effective at delaying tumour recurrence by 12 (p < 0.05) or 21 days (p < 0.001), respectively, by reducing the number of CSCs. The potential mechanism of action, in addition to CD44, involves downregulation of DLL4 and Notch4. CONCLUSION: This study demonstrates, for the first time, the pre-clinical activity of novel systemic anti-cancer therapeutic peptides, ALM201 and AD-01, in the metastatic setting, and highlights their impact on endocrine therapy resistant CSCs; both areas of unmet clinical need.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Imunofilinas/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Peptídeos/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Mama/patologia , Neoplasias da Mama/patologia , Proteínas de Ligação ao Cálcio , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imunofilinas/uso terapêutico , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos SCID , Recidiva Local de Neoplasia/prevenção & controle , Células-Tronco Neoplásicas/patologia , Peptídeos/uso terapêutico , Receptor Notch4/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas de Ligação a Tacrolimo , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Clin Cancer Res ; 19(14): 3881-93, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23741069

RESUMO

PURPOSE: FK506-binding protein like (FKBPL) and its peptide derivative, AD-01, have already shown tumor growth inhibition and CD44-dependent antiangiogenic activity. Here, we explore the ability of AD-01 to target CD44-positive breast cancer stem cells (BCSC). EXPERIMENTAL DESIGN: Mammosphere assays and flow cytometry were used to analyze the effect of FKBPL overexpression/knockdown and AD-01 treatment ± other anticancer agents on BCSCs using breast cancer cell lines (MCF-7/MDA-231/ZR-75), primary patient samples, and xenografts. Delays in tumor initiation were evaluated in vivo. The anti-stem cell mechanisms were determined using clonogenic assays, quantitative PCR (qPCR), and immunofluorescence. RESULTS: AD-01 treatment was highly effective at inhibiting the BCSC population by reducing mammosphere-forming efficiency and ESA(+)/CD44(+)/CD24(-) or aldehyde dehydrogenase (ALDH)(+) cell subpopulations in vitro and tumor initiation in vivo. The ability of AD-01 to inhibit the self-renewal capacity of BCSCs was confirmed; mammospheres were completely eradicated by the third generation. The mechanism seems to be due to AD-01-mediated BCSC differentiation shown by a significant decrease in the number of holoclones and an associated increase in meroclones/paraclones; the stem cell markers, Nanog, Oct4, and Sox2, were also significantly reduced. Furthermore, we showed additive inhibitory effects when AD-01 was combined with the Notch inhibitor, DAPT. AD-01 was also able to abrogate a chemo- and radiotherapy-induced enrichment in BCSCs. Finally, FKBPL knockdown led to an increase in Nanog/Oct4/Sox2 and an increase in BCSCs, highlighting a role for endogenous FKBPL in stem cell signaling. CONCLUSIONS: AD-01 has dual antiangiogenic and anti-BCSC activity, which will be advantageous as this agent enters clinical trial.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Receptores de Hialuronatos/metabolismo , Imunofilinas/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Imunofilinas/metabolismo , Células MCF-7 , Camundongos , Camundongos SCID , Tolerância a Radiação , Receptores Notch/antagonistas & inibidores , Receptores Notch/metabolismo , Transdução de Sinais , Esferoides Celulares/efeitos dos fármacos , Proteínas de Ligação a Tacrolimo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Biochim Biophys Acta ; 1833(3): 652-62, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23228564

RESUMO

Immunophilins are FK506-binding proteins that have been involved in the regulation of calcium homeostasis, either by modulating Ca(2+) channels located in the plasma membrane or in the rough endoplasmic reticulum (RE). We have investigated whether immunophilins would participate in the regulation of stored-operated Ca(2+) entry (SOCE) in human platelets and MEG 01. Both cell types were loaded with fura-2 for determining cytosolic calcium concentration changes ([Ca(2+)](c)), or stimulated and fixed to evaluate the protein interaction profile by performing immunoprecipitation and western blotting. We have found that incubation of platelets with FK506 increases Ca(2+) mobilization. Thapsigargin (TG)-evoked, Thr-evoked SOCE and TG-evoked Mn(2+) entry resulted in significant reduction by treatment of platelets with immunophilin antagonists. We confirmed by immunoprecipitation that immunophilins interact with transient receptor potential channel 1 (TRPC1) and Orai1 in human platelets. FK506 and rapamycin reduced the association between TRPC1 and Orai1 with FK506 binding protein (52) (FKBP52) in human platelets, and between TRPC1 and the type II IP(3)R, which association is known to be crucial for the maintenance of SOCE in human platelets. FKBP52 role in SOCE activation was confirmed by silencing FKBP52 using SiRNA FKBP52 in MEG 01 as demonstrated by single cell configuration imaging technique. TRPC1 silencing and depletion of cell of TRPC1 and FKBP52 simultaneously, impair activation of SOCE evoked by TG in MEG 01. Finally, in MEG 01 incubated with FK506 we observed a reduction in TRPC1/FKBP52 coupling, and similarly, FKBP52 silencing reduced the association between IP3R type II and TRPC1 during SOCE. All together, these results demonstrate that immunophilins participate in the regulation of SOCE in human platelets.


Assuntos
Plaquetas/metabolismo , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Células Progenitoras de Megacariócitos/metabolismo , Canais de Cátion TRPC/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Plaquetas/efeitos dos fármacos , Western Blotting , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Fura-2/metabolismo , Humanos , Imunofilinas/farmacologia , Imunoprecipitação , Imunossupressores/farmacologia , Transporte de Íons/efeitos dos fármacos , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/efeitos dos fármacos , Proteína ORAI1 , RNA Interferente Pequeno/genética , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/genética , Tacrolimo/farmacologia , Proteínas de Ligação a Tacrolimo/antagonistas & inibidores , Proteínas de Ligação a Tacrolimo/genética , Tapsigargina/farmacologia
6.
J Neurosci Res ; 90(7): 1413-23, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22431363

RESUMO

Stroke is the leading cause of disability in the industrialized world, and the development of pharmacologic strategies to promote poststroke recovery is of paramount importance. GM1485, a nonimmunosuppressive immunophilin ligand, promotes regeneration of multiple cell types following injury. In the present study, we evaluated the effect of GM1485 treatment on functional recovery and neurogenesis following rat stroke. Transient cerebral ischemia was induced in rats receiving daily GM1485 (5 mg/kg) beginning 24 hr postischemia and continuing for a total of 6 weeks. Neurological function was evaluated over this period using a battery of neurobehavioral tests, and immunostaining for stem-cell markers was performed following animal sacrifice. An in vitro model of oxidative stress was also employed to evaluate the ability of GM1485 to mediate stem-cell-like induction and plasticity. GM1485-treated rats demonstrated improved neurological function as well as increased Sox2(+) cells in the ipsilateral SVZ and striatum relative to vehicle-treated rats. Additionally, GM1485-treated fibroblasts subjected to oxidative stress were reprogrammed to a stem-cell-like phenotype and were able to differentiate down a neuronal lineage. These data demonstrate that GM1485 administration improves neurological function and is consistent with an upregulation of endogenous neurogenesis following stroke in rats. Further experiments are necessary to characterize the molecular pathways involved in these processes.


Assuntos
Imunofilinas/farmacologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fatores de Crescimento Neural/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Recuperação de Função Fisiológica/efeitos dos fármacos , Tacrolimo/análogos & derivados , Animais , Modelos Animais de Doenças , Imunofilinas/uso terapêutico , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Infusões Intravenosas , Ligantes , Masculino , Fatores de Crescimento Neural/uso terapêutico , Regeneração Nervosa/fisiologia , Cultura Primária de Células , Ratos , Ratos Wistar , Recuperação de Função Fisiológica/fisiologia , Tacrolimo/farmacologia , Tacrolimo/uso terapêutico , Resultado do Tratamento
7.
Clin Cancer Res ; 17(5): 1044-56, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21364036

RESUMO

PURPOSE: Antiangiogenic therapies can be an important adjunct to the management of many malignancies. Here we investigated a novel protein, FKBPL, and peptide derivative for their antiangiogenic activity and mechanism of action. EXPERIMENTAL DESIGN: Recombinant FKBPL (rFKBPL) and its peptide derivative were assessed in a range of human microvascular endothelial cell (HMEC-1) assays in vitro. Their ability to inhibit proliferation, migration, and Matrigel-dependent tubule formation was determined. They were further evaluated in an ex vivo rat model of neovascularization and in two in vivo mouse models of angiogenesis, that is, the sponge implantation and the intravital microscopy models. Antitumor efficacy was determined in two human tumor xenograft models grown in severe compromised immunodeficient (SCID) mice. Finally, the dependence of peptide on CD44 was determined using a CD44-targeted siRNA approach or in cell lines of differing CD44 status. RESULTS: rFKBPL inhibited endothelial cell migration, tubule formation, and microvessel formation in vitro and in vivo. The region responsible for FKBPL's antiangiogenic activity was identified, and a 24-amino acid peptide (AD-01) spanning this sequence was synthesized. It was potently antiangiogenic and inhibited growth in two human tumor xenograft models (DU145 and MDA-231) when administered systemically, either on its own or in combination with docetaxel. The antiangiogenic activity of FKBPL and AD-01 was dependent on the cell-surface receptor CD44, and signaling downstream of this receptor promoted an antimigratory phenotype. CONCLUSION: FKBPL and its peptide derivative AD-01 have potent antiangiogenic activity. Thus, these agents offer the potential of an attractive new approach to antiangiogenic therapy.


Assuntos
Inibidores da Angiogênese/farmacologia , Imunofilinas/uso terapêutico , Neoplasias/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Inibidores da Angiogênese/química , Inibidores da Angiogênese/uso terapêutico , Animais , Western Blotting , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Docetaxel , Células Endoteliais/efeitos dos fármacos , Receptores de Hialuronatos/genética , Imunofilinas/química , Imunofilinas/farmacologia , Imunoprecipitação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Neoplasias/tratamento farmacológico , Fragmentos de Peptídeos/uso terapêutico , RNA Interferente Pequeno/genética , Ratos , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas de Ligação a Tacrolimo , Taxoides/farmacologia , Taxoides/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Biotechnol Lett ; 32(9): 1333-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20443046

RESUMO

Immunophilin ligand FK506 has been treated as adjunct therapy for nerve repair due to its potent neurotrophic and neuroprotective actions. It was hypothesized that FK506 releasing from biodegradable chitosan guide provided better nerve regenerative response than the guide with no FK506. The drug was entrapped in the semi-permeable wall of chitosan guide with the drug-loading of 647 microg/g. Rat sciatic nerve defect model treated with FK506-releasing chitosan guide showed more mature appearance of myelinated fibers 8 weeks after surgery; furthermore, the motor functional reinnervation occurred, the amplitude and velocity of compound muscle action potentials reached 60% and 73% with respect to the normal. Thus, FK506-releasing chitosan guide should be acted as a long-lasting delivery device of immunosuppressive and neuroregenerative agent for peripheral nerve repair.


Assuntos
Quitosana/metabolismo , Sistemas de Liberação de Medicamentos , Imunofilinas/farmacologia , Fator de Crescimento Neural/farmacologia , Regeneração Nervosa , Nervo Isquiático/efeitos dos fármacos , Tacrolimo/farmacologia , Animais , Ratos , Recuperação de Função Fisiológica/efeitos dos fármacos , Nervo Isquiático/lesões , Nervo Isquiático/fisiologia
9.
J Sex Med ; 6(7): 1914-23, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19453873

RESUMO

INTRODUCTION: Immunophilin ligands function by binding to receptor proteins such as FK506 binding proteins (FKBPs). FKBPs are studied for their roles in neuroprotection. AIM: Compare the effect of FK506 (FK) and rapamycin (RAP) on erectile function (EF) recovery and FKBP expressions in penis and major pelvic ganglion (MPG) after cavernous nerve (CN) injury. METHODS: Adult male rats were divided into four groups: sham surgery (CN exposure only) + vehicle; bilateral CN injury (BCNI; bilateral crush, 3 minutes with hemostat clamp) + vehicle; BCNI + FK (5 mg/kg/day, 5 days, sc); and BCNI + RAP (2 mg/kg/day, 5 days, sc). At both 24 hours (Day 1) or 1 week (Day 7) after BCNI, EF was assessed by intracavernosal pressure measurement and FKBPs 12, 38, 52, and 65 expressions were evaluated by Western blot analysis in collected penises and MPGs. MAIN OUTCOME MEASURES: EF and change in protein expressions of FKBPs in the rat penis and MPG after BCNI with and without immunophilin ligand treatment. RESULTS: Both FK- and RAP-treated rats had preserved EF compared with vehicle-treated rats after BCNI. FKBPs changed variably following injury and treatment. In particular, in the penis at Day 1, FKBP 38 expression was decreased after BCNI and both FK and RAP attenuated this decrease. In MPG at Day 1, FKBP 38 expression was also decreased after BCNI and FK attenuated the decrease, while at Day 7, FKBP 38 expression was still decreased and RAP attenuated the decrease. Also, in the penis at Day 1, FKBP 65 expression decreased after BCNI and FK attenuated the decrease. In the MPG, FKBP 65 expression increased at both Days 1 and 7 with FK treatment. CONCLUSIONS: Improved EF after BCNI, as shown with RAP, further suggests a role of immunophilin ligands as a protective therapy of CN injury associated erectile dysfunction. Our findings also suggest that select FKBPs, such as FKBP 38 and FKBP 65, may mediate these effects.


Assuntos
Imunossupressores/farmacologia , Fármacos Neuroprotetores/farmacologia , Pênis/efeitos dos fármacos , Sirolimo/farmacologia , Proteínas de Ligação a Tacrolimo/farmacologia , Tacrolimo/farmacologia , Animais , Vias Autônomas , Gânglios/efeitos dos fármacos , Gânglios/lesões , Imunofilinas/farmacologia , Masculino , Modelos Animais , Pênis/inervação , Ratos
10.
Eur J Pharmacol ; 591(1-3): 147-52, 2008 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-18602914

RESUMO

Injured spinal cord axons fail to regenerate in part due to a lack of trophic support. While various methods for replacing neurotrophins have been pursued, clinical uses of these methods face significant barriers. FK1706, a non-immunosuppressant neurophilin ligand, potentiates nerve growth factor signaling, suggesting therapeutic potential for functional deficits following spinal cord injury. Here, we demonstrate that FK1706 significantly improves behavioral outcomes in animal models of spinal cord hemisection and contusion injuries in rats. Furthermore, we show that FK1706 is effective even if administration is delayed until 1 week after injury, suggesting that FK1706 has a reasonable therapeutic time-window. Morphological analysis of injured axons in the dorsal corticospinal tract showed an increase in the radius and perimeter of stained axons, which were reduced by FK1706 treatment, suggesting that axonal swelling and retraction balls observed in injured spinal cord were improved by the neurotrophic effect of FK1706. Taken together, FK1706 improves both behavioral motor function and the underlying morphological changes, suggesting that FK1706 may have therapeutic potential in meeting the significant unmet needs in spinal cord injury.


Assuntos
Regeneração Nervosa/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Traumatismos da Medula Espinal/tratamento farmacológico , Tacrolimo/análogos & derivados , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Modelos Animais de Doenças , Imunofilinas/farmacologia , Masculino , Fator de Crescimento Neural/efeitos dos fármacos , Fator de Crescimento Neural/metabolismo , Tratos Piramidais/metabolismo , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Traumatismos da Medula Espinal/fisiopatologia , Tacrolimo/farmacologia , Fatores de Tempo
11.
J Neuroimmune Pharmacol ; 2(1): 49-57, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18040826

RESUMO

HIV infection results in a neurodegenerative disorder for which currently there is no effective therapy available. Currently, available antiretroviral therapy has no impact on the production of early regulatory HIV proteins once the virus is integrated. Of these proteins, Tat was shown to be toxic to neurons. We, thus, used an in vitro neuronal culture system to determine if immunophilin ligands could protect against Tat-induced neurotoxicity. We found that GPI 1046 had potent neuroprotective effects in this model. The compound was able to protect the neurons even though it only partially obliterated Tat-induced oxidative stress in neurons, suggesting that other mechanisms may be important in mediating its neuroprotective effect. Furthermore, GPI 1046 showed inhibition of HIV replication and Tat-mediated long terminal repeat (LTR) activation suggesting that this class of compounds may be worthy of further exploration as a potential treatment for HIV dementia.


Assuntos
Antirretrovirais/farmacologia , Imunofilinas/metabolismo , Fármacos Neuroprotetores/farmacologia , Pirrolidinas/farmacologia , Animais , Antirretrovirais/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Células Cultivadas , Produtos do Gene tat , Infecções por HIV/tratamento farmacológico , Infecções por HIV/patologia , Humanos , Imunofilinas/farmacologia , Imunofilinas/uso terapêutico , Células Jurkat , Ligantes , Fármacos Neuroprotetores/uso terapêutico , Pirrolidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia
12.
J Pharmacol Sci ; 102(1): 77-87, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16990701

RESUMO

In the present study, we investigated whether the protective effect of FK506 and cyclosporin A (CsA) against in vitro ischemic injury of astrocytes might be mediated through attenuation of cytosolic isoform of phospholipase A(2) (cPLA(2)) expression and activity as well as inhibition of arachidonic acid (AA) release. On the 21st day in vitro, cultures of rat astrocytes were subjected to ischemia-simulating conditions (combined oxygen glucose deprivation) for 8 h and exposed to FK506 (10 - 1,000 nM) and CsA (0.25 - 10 microM). Obtained data suggest the cross-talk between the action of 0.25 - 10 microM CsA as well as 1 microM FK506 on calcineurin (CaN) and cPLA(2) in anti-apoptotic signal transduction pathways. Moreover, we have shown that immunosuppressants at these concentrations protected glial cells against ischemia-induced apoptosis through the increase of cell viability, mitochondrial function restoration, and attenuation of oxidative stress. Finally, in our study, low concentrations of FK506 (10 and 100 nM) exerted limited effects on the assessed parameters. Our findings document a key role either for CaN or cPLA(2) expression attenuation and AA release inhibition in the antiapoptotic effect of FK506 and CsA in ischemic astrocytes.


Assuntos
Apoptose/efeitos dos fármacos , Ácido Araquidônico/metabolismo , Astrócitos/efeitos dos fármacos , Isquemia Encefálica/patologia , Ciclosporina/farmacologia , Citosol/enzimologia , Citosol/metabolismo , Imunossupressores/farmacologia , Fosfolipases A/metabolismo , Tacrolimo/farmacologia , Animais , Animais Recém-Nascidos , Antioxidantes/metabolismo , Western Blotting , Calcineurina/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Imunofilinas/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fosfolipases A2 , Ratos , Ratos Wistar , Receptor Cross-Talk/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
13.
Cell ; 122(6): 861-73, 2005 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-16179256

RESUMO

The meiotic recombination checkpoint delays gamete precursors in G2 until DNA breaks created during recombination are repaired and chromosome structure has been restored. Here, we show that the FK506 binding protein Fpr3 prevents premature adaptation to damage and thus serves to maintain recombination checkpoint activity. Impaired checkpoint function is observed both in cells lacking FPR3 and in cells treated with rapamycin, a small molecule inhibitor that binds to the proline isomerase (PPIase) domain of Fpr3. FPR3 functions in the checkpoint through controlling protein phosphatase 1 (PP1). Fpr3 interacts with PP1 through its PPIase domain, regulates PP1 localization, and counteracts the activity of PP1 in vivo. Our findings define a branch of the recombination checkpoint involved in the adaptation to persistent chromosomal damage and a critical function for FK506 binding proteins during meiosis.


Assuntos
Imunofilinas/fisiologia , Meiose , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/fisiologia , Recombinação Genética/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Dano ao DNA , Regulação da Expressão Gênica , Imunofilinas/genética , Imunofilinas/farmacologia , Peptidilprolil Isomerase/antagonistas & inibidores , Peptidilprolil Isomerase/genética , Fosfoproteínas Fosfatases/genética , Mutação Puntual , Proteína Fosfatase 1 , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/fisiologia , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/farmacologia , Sirolimo/farmacologia , Fatores de Tempo
14.
Neurotoxicology ; 26(3): 373-84, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15935209

RESUMO

The aim of the present study was to test the hypothesis that exposure of astrocytes depleted of glutathione (GSH) to simulated ischemia conditions in vitro and treated with immunosuppressant immunophilin ligands (cyclosporin A (CsA) and FK506) can increase intracellular GSH levels and that such mechanism may be responsible, at least in part, for their protective effects. In addition, we also compared the antioxidant properties of these immunosuppressants with N-acetylcysteine (NAC), a precursor of GSH synthesis. GSH depletion was induced by 24 h pretreatment with L-buthionine sulfoximine (BSO). Cultures of rat astrocytes were exposed to CsA (1-50 microM) and FK506 (1-1000 nM) and NAC (100 or 200 microM). We examined the effects of these compounds on apoptosis, cell viability, reactive oxygen species production and GSH content. Our study demonstrated that toxicity of simulated ischemia conditions were enhanced when intracellular GSH was depleted, and immunosuppressants (especially 100 nM FK506 and 10 microM CsA) effectively prevented ischemia toxicity in GSH depleted astrocytes. In addition, we have shown that interfering with the generation of GSH and attenuation, the rise of oxidative stress level by NAC may be a powerful tool for prevention of ischemia-induced glial cell damage.


Assuntos
Acetilcisteína/farmacologia , Astrócitos/efeitos dos fármacos , Isquemia Encefálica/patologia , Sequestradores de Radicais Livres/farmacologia , Glutationa/fisiologia , Imunofilinas/farmacologia , Imunossupressores/farmacologia , Animais , Apoptose/efeitos dos fármacos , Benzimidazóis , Separação Celular , Células Cultivadas , Ciclosporina/farmacologia , Corantes Fluorescentes , Glutationa/metabolismo , Ligantes , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Tacrolimo/farmacologia , Sais de Tetrazólio , Tiazóis
15.
J Steroid Biochem Mol Biol ; 94(1-3): 71-81, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15862952

RESUMO

In MCF-7 (estrogen receptor (ER)+) and in MDA-MB-231 (ER-) cells stably transfected with either estrogen receptor alpha (ERalpha) or beta (ERbeta) subtype (MDA-MB-231 stably transfected with the mouse ERalpha cDNA (MERA) and MDA-MB-231 stably transfected with the human ERbeta cDNA (HERB), respectively) N-term heat shock protein of 90kDa (hsp90) ligands (geldanamycin and radicicol) and C-term hsp90 ligands (novobiocin) decrease the basal and estradiol (E(2))-induced transcription activity of ER on an estrogen responsive element (ERE)-LUC reporter construct concomitantly with or 1h after E(2) treatment. All hsp90 ligands induced an E(2)- and MG132-inhibited decrease of both ER cell content. However, the kinetics of these degradations are slower than those induced by the selective estrogen receptor down-regulator RU 58668 (RU). This suggests that inhibition of the hsp90 ATPase activity targets both ERs to the 26S proteasome and that hsp90 interacts with both ER subtypes. Rapamycin (Rapa) and cyclosporin A (CsA), ligands of immunophilins FK506 binding protein (FKBP52) and cyclophilin of 40kDa (CYP40) interacting in separate ER-hsp90 complexes, both induced a proteasomal-mediated degradation of ERs but not of their cognate immunophilin. Moreover, they also decrease the E(2)-induced luciferase transcription but weaker than RU and hsp90 ligands. Fluorescence activated cell sorter (FACS) analysis revealed a blockade of cell progression by RU and 4-hydroxy-tamoxifen at the G(1) phase of the cell cycle and an induction of apoptosis in MCF-7 cells. Rapa and mainly CsA (but not FK506) and hsp90 ligands promote by their own apoptosis in MCF-7, in MERA, and in HERB cells and in MDA-MB-231 ER-null cells. These data suggest that (1) hsp90, as for all steroid receptors, acts as a molecular chaperone for ERbeta; (2) ER-ligands (except tamoxifen), hsp90- and immunophilin-ligands (except FK506) target the two ER subtypes to a proteasome-mediated proteolysis via different signalling pathways; (3) hsp90- and immunophilin-ligands Rapa and CsA, alone or in association with anti-estrogens such as RU, may constitute a potential therapeutic strategy for breast cancer treatment.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/metabolismo , Imunofilinas/farmacologia , Neoplasias da Mama , Linhagem Celular Tumoral , DNA Complementar/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Proteínas de Choque Térmico HSP90/genética , Humanos , Cinética , Ligantes , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional , Transfecção
16.
Urology ; 65(5): 1003-7, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15882753

RESUMO

OBJECTIVES: Nonimmunosuppressant immunophilin ligands such as GPI-1046 have gained interest recently for their clinical potential to reduce the extent of injury sustained by penile innervation during radical prostatectomy for prostate cancer treatment and thereby improve erectile function recovery postoperatively. Preclinical studies in rat animal models of cavernous nerve injury have demonstrated that immunophilin ligands exert both neuroprotective and neuroregenerative effects on physiologic erection and on the morphology of erectile tissue and penile innervation. Before establishing their clinical roles, however, it would be useful to evaluate whether they exert mitogenic effects on malignant prostate cells. METHODS: Human prostate cancer cell lines (LAPC-4, CWR22Rv1, LNCaP, and PC-3) were treated in vitro with 0 to 10 microM of the prototypical nonimmunosuppressant ligand GPI-1046 in 1% fetal bovine serum (FBS)-containing (ie, low growth) media, and their growth was assayed during a 7-day period using spectrophotometric cell counting. The results were normalized for comparison to the maximal growth produced in 10% FBS-containing media. RESULTS: All four prostate cancer cell lines grew maximally in 10% FBS-containing media, and this growth was reduced by more than twofold (P <0.05) when the cells were maintained in 1% FBS-containing media. The addition of 10 microM or less GPI-1046 to the cells in 1% FBS-containing media did not exert statistically significant mitogenic effects on human prostate cancer cells in vitro. CONCLUSIONS: These data contribute toward allaying concerns that the use of nonimmunosuppressant immunophilin ligand therapy for the improvement of erectile function recovery after radical prostatectomy will promote recurrent prostate cancer.


Assuntos
Imunofilinas/farmacologia , Neoplasias da Próstata/patologia , Pirrolidinas/farmacologia , Divisão Celular/efeitos dos fármacos , Humanos , Ligantes , Masculino , Fármacos Neuroprotetores/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos
17.
Eur J Pharmacol ; 509(1): 11-9, 2005 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-15713424

RESUMO

Immunophilin ligands are neuroregenerative agents, characterized by binding to FK506 binding proteins (FKBPs), which stimulate recovery of neurons in a variety of injury paradigms. Here we report the discovery of a novel, non-immunosuppressive immunophilin ligand, FK1706. FK1706, a derivative of FK506, showed similarly high affinity for two FKBP subtypes, FKBP-12 and FKBP-52, but inhibited T-cell proliferation and interleukin-2 cytokine production with much lower potency and efficacy than FK506. FK1706 (0.1 to 10 nM) significantly potentiated nerve growth factor (NGF)-induced neurite outgrowth in SH-SY5Y cells, as did FK506. This neurite potentiation could be blocked by an anti-FKBP-52 antibody, as well as by specific pharmacological inhibitors of phospholipase C (PLC), phosphatidylinositol 3-kinase (PI3K), and the Ras/Raf/Mitogen-Activated Protein Kinase (MAPK) signaling pathway. FK1706 also potentiated NGF-induced MAPK activation, with a similar dose-dependency to that necessary for potentiating neurite outgrowth. Taken together, these data suggest that FK1706 is a non-immunosuppressive immunophilin ligand with significant neurotrophic effects, putatively mediated via FKBP-52 and the Ras/Raf/MAPK signaling pathway, and therefore that FK1706 may have therapeutic potential in a variety of neurological disorders.


Assuntos
Imunofilinas/farmacologia , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/fisiologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Imunofilinas/química , Imunofilinas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Crescimento Neural/antagonistas & inibidores , Receptor trkB/metabolismo , Transdução de Sinais/fisiologia , Tacrolimo/análogos & derivados , Tacrolimo/química , Tacrolimo/imunologia , Tacrolimo/metabolismo , Tacrolimo/farmacologia , Proteína 1A de Ligação a Tacrolimo/química , Proteína 1A de Ligação a Tacrolimo/metabolismo , Proteína 1A de Ligação a Tacrolimo/farmacologia , Trítio
18.
Neuropharmacology ; 48(3): 391-7, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15721171

RESUMO

Repeated use of methamphetamine (MAP) is known to cause neurotoxicity in the dopaminergic neurons of the striatum. Recently, we reported that FK506, a calcineurin inhibitor and immunosuppressive agent, could attenuate acute behavioral changes and the development of sensitization after administration of MAP. In this study, we investigated the effects of FK506 on the neurotoxicity in the dopaminergic neurons induced by repeated administration of MAP. BALB/c mice were injected subcutaneously (s.c.) with vehicle (10 ml/kg) or MAP (4 mg/kg) four times every 2h. Vehicle (10 ml/kg) or FK506 (0.1, 0.3, 1 or 3 mg/kg i.p.) was administered 15 min before the first MAP administration. Three days later, we assessed the contents of dopamine (DA) and its major metabolite, 3,4-dihydroxyphenylacetic acid (DOPAC), in the mouse striatum using high-performance liquid chromatography (HPLC). We also examined the immunohistochemistry of dopamine transporter (DAT) and tyrosine hydroxylase (TH) in the mouse brain. Repeated administration of MAP decreased significantly the contents of DA and DOPAC in the mouse striatum, and pretreatment with FK506 inhibited significantly the reduction of DA and DOPAC in the mouse brain by repeated administration of MAP. Furthermore, repeated administration of MAP decreased significantly the immunoreactivity of DAT and TH in the striatum as compared to controls. Pretreatment with FK506 (3 mg/kg) attenuated significantly the reduction of DAT and TH immunoreactivity after repeated administration of MAP. These results suggest that FK506 shows protective effects on the MAP-induced neurotoxicity in the dopaminergic neurons of the mouse striatum.


Assuntos
Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , Metanfetamina/toxicidade , Fármacos Neuroprotetores/farmacologia , Tacrolimo/farmacologia , Animais , Corpo Estriado/metabolismo , Relação Dose-Resposta a Droga , Imunofilinas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C
19.
J Neurosci Res ; 77(3): 367-77, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15248293

RESUMO

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) in which demyelination and axonal loss result in permanent neurologic disability. We examined the neuroprotective property of the immunosuppressant FK506 (tacrolimus), FK1706 (a nonimmunosuppressant FK506 derivative) and cyclosporin A (CsA) in a chronic relapsing experimental autoimmune encephalomyelitis (EAE) model of MS. Female SJL/J mice were immunized by subcutaneous (s.c.) injection with proteolipid protein 139-151 peptide in complete Freund's adjuvant. At the onset of paralysis, 12-14 days after immunization, mice received daily s.c. injections of FK506 (0.2, 1, and 5 mg/kg), FK1706 (5 mg/kg), CsA (2, 10, and 50 mg/kg), saline or vehicle (30% dimethylsulfoxide) for 30 days. FK506 (at a dose of 5 mg/kg) reduced the severity of the initial disease and suppressed relapses. FK1706 did not significantly alter the clinical course and CsA (at a dose of 50 mg/kg) lessened the severity of the initial episode of EAE but did not alter relapses. In the thoracic spinal cord, FK506 (5 mg/kg), FK1706 (5 mg/kg), and CsA (50 mg/kg) significantly (P < 0.001) reduced the extent of damage in the dorsal, lateral, and ventral white matter by a mean of up to 95, 68, and 30%, respectively. A nonimmunosuppressant dose of FK506 (0.2 mg/kg) also significantly (P < 0.001) reduced the extent of damage in the spinal cord by a mean of up to 45%. Other dosages of these compounds were ineffective. FK506 markedly protects against demyelination and axonal loss in this MS model through immunosuppression and neuroprotection.


Assuntos
Axônios/efeitos dos fármacos , Encefalomielite Autoimune Experimental/tratamento farmacológico , Esclerose Múltipla/tratamento farmacológico , Bainha de Mielina/efeitos dos fármacos , Tacrolimo/análogos & derivados , Tacrolimo/uso terapêutico , Animais , Axônios/patologia , Relação Dose-Resposta a Droga , Encefalomielite Autoimune Experimental/patologia , Feminino , Imunofilinas/farmacologia , Imunofilinas/uso terapêutico , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Camundongos , Esclerose Múltipla/patologia , Bainha de Mielina/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Tacrolimo/farmacologia
20.
Artigo em Japonês | MEDLINE | ID: mdl-15164613

RESUMO

Non-immunosuppressive immunophilin ligands (NI-IPLs) are attracting attention as new candidate drugs for neuroprotection and/or neurorestoration, particularly since they do not have the adverse effects of immunosuppressants. However, it is not yet enough to understand that NI-IPLs are useful drugs for treating neurological disorders. In particular, the molecular mechanism of NI-IPL activity in target cells in the brain remains obscure. In this review, we focused on the molecular basis of the neuroprotective properties of IPLs. Our findings suggest that IPLs have neuroprotective effects mediated by multiple beneficial properties such as a glutathione (GSH)-activating effect, a neurotrophic factor (NTF)-activating effect, and an anti-apoptotic effect, but not by an immunosuppressive effect, both in cell cultures and in vivo. In particular, the GSH-activating effect and the NTF-activating effect of NI-IPLs may be essential to the expression of their neuroprotective properties. Thus, NI-IPLs might have a potentially beneficial effect by ameliorating neurological disorders, since they do not cause serious side effects such as immune deficiency.


Assuntos
Imunofilinas/farmacologia , Fármacos Neuroprotetores , Animais , Ciclosporina/metabolismo , Glutationa/metabolismo , Humanos , Imunofilinas/metabolismo , Imunofilinas/uso terapêutico , Imunossupressores/metabolismo , Ligantes , Fatores de Crescimento Neural/metabolismo , Doenças do Sistema Nervoso/tratamento farmacológico , Tacrolimo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...