Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Biosci (Landmark Ed) ; 29(3): 109, 2024 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-38538266

RESUMO

BACKGROUND: Severe neurological condition like Alzheimer's disease (AD) has a significantly negative impact on families and society, wherein there is no proven cure. As one of the principal active constituents of Achyranthes bidentata Blume, ecdysterone (ECR) has demonstrated antioxidant and cognitive dysfunction improvement effects. Nonetheless, the mechanism underlying the improvement of cognitive dysfunction by ECR remains unclear. This study sought to ascertain whether ECR may allebviate cognitive impairment by reducing oxidative stress via activation of the nuclear factor erythroid-2-related factor-2 (Nrf2) antioxidant system through Akt/GSK3ß pathway. METHODS: In terms of the experimental procedure, we determined the neuroprotective benefits of ECR in vivo via a cognitive impairment model of senescence-accelerated mouse prone 8 (SAMP8), we performed procedures such as behavioral testing, biochemical assaying, Nissl and TUNEL stainings, as well as flow cytometry, immunohistochemistry and western blotting. Furthermore, we investigated the underlying mechanistic action of ECR by activating PC12 cells with ß-amyloid peptide fragment 25-35 (Aß25-35). RESULTS: In vivo studies showed that ECR effectively improved cognitive impairment in SAMP8 via enhancement of learning and memory capabilities, but decreased oxidative stress, apoptosis and neuronal damage in the hippocampus. During the in vitro study, we observed that ECR dose-dependently reduced the oxidative stress and apoptosis that were induced in PC12 cells by Aß25-35. Additionally, the use of Akt inhibitors further established the potential of ECR to control Nrf2 through activation of the Akt/GSK3ß pathway and protect the PC12 cells from Aß25-35 induced damage. CONCLUSIONS: These findings offer proof that ECR reduces cognitive impairment by triggering the Nrf2 antioxidant system via the Akt/GSK3ß pathway and offer fresh information on ECR's potential as a promising therapeutic development candidate for AD.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Fármacos Neuroprotetores , Humanos , Ratos , Camundongos , Animais , Glicogênio Sintase Quinase 3 beta/metabolismo , Antioxidantes/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Ecdisterona/farmacologia , Ecdisterona/uso terapêutico , Estresse Oxidativo , Transdução de Sinais , Peptídeos beta-Amiloides/toxicidade , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/tratamento farmacológico , Disfunção Cognitiva/tratamento farmacológico , Cognição , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico
2.
Trials ; 22(1): 42, 2021 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-33430924

RESUMO

OBJECTIVES: As of December, 1st, 2020, coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2, resulted in more than 1 472 917 deaths worldwide and death toll is still increasing exponentially. Many COVID-19 infected people are asymptomatic or experience moderate symptoms and recover without medical intervention. However, older people and those with comorbid hypertension, diabetes, obesity, or heart disease are at higher risk of mortality. Because current therapeutic options for COVID-19 patients are limited specifically for this elderly population at risk, Biophytis is developing BIO101 (20-hydroxyecdysone, a Mas receptor activator) as a new treatment option for managing patients with SARS-CoV-2 infection at the severe stage. The angiotensin converting enzyme 2 (ACE2) serves as a receptor for SARS-CoV-2. Interaction between ACE2 and SARS-CoV2 spike protein seems to alter the function of ACE2, a key player in the renin-angiotensin system (RAS). The clinical picture of COVID-19 includes acute respiratory distress syndrome (ARDS), cardiomyopathy, multiorgan dysfunction and shock, all of which might result from an imbalance of the RAS. We propose that RAS balance could be restored in COVID-19 patients through MasR activation downstream of ACE2 activity, with 20-hydroxyecdysone (BIO101) a non-peptidic Mas receptor (MasR) activator. Indeed, MasR activation by 20-hydroxyecdysone harbours anti-inflammatory, anti-thrombotic, and anti-fibrotic properties. BIO101, a 97% pharmaceutical grade 20-hydroxyecdysone could then offer a new therapeutic option by improving the respiratory function and ultimately promoting survival in COVID-19 patients that develop severe forms of this devastating disease. Therefore, the objective of this COVA study is to evaluate the safety and efficacy of BIO101, whose active principle is 20-hydroxyecdysone, in COVID-19 patients with severe pneumonia. TRIAL DESIGN: Randomized, double-blind, placebo-controlled, multi-centre, group sequential and adaptive which will be conducted in 2 parts. Part 1: Ascertain the safety and tolerability of BIO101 and obtain preliminary indication of the activity of BIO101, in preventing respiratory deterioration in the target population Part 2: Re-assessment of the sample size needed for the confirmatory part 2 and confirmation of the effect of BIO101 observed in part 1 in the target population. The study is designed as group sequential to allow an efficient run-through, from obtaining an early indication of activity to a final confirmation. And adaptive - to allow accumulation of early data and adapt sample size in part 2 in order to inform the final design of the confirmatory part of the trial. PARTICIPANTS: Inclusion criteria 1. Age: 45 and above 2. A confirmed diagnosis of COVID-19 infection, within the last 14 days, prior to randomization, as determined by PCR or other approved commercial or public health assay, in a specimen as specified by the test used. 3. Hospitalized, in observation or planned to be hospitalized due to COVID-19 infection symptoms with anticipated hospitalization duration ≥3 days 4. With evidence of pneumonia based on all of the following: a. Clinical findings on a physical examination b. Respiratory symptoms developed within the past 7 days 5. With evidence of respiratory decompensation that started not more than 4 days before start of study medication and present at screening, meeting one of the following criteria, as assessed by healthcare staff: a. Tachypnea: ≥25 breaths per minute b. Arterial oxygen saturation ≤92% c. A special note should be made if there is suspicion of COVID-19-related myocarditis or pericarditis, as the presence of these is a stratification criterion 6. Without a significant deterioration in liver function tests: a. ALT and AST ≤ 5x upper limit of normal (ULN) b. Gamma-glutamyl transferase (GGT) ≤ 5x ULN c. Total bilirubin ≤ 5×ULN 7. Willing to participate and able to sign an informed consent form (ICF). Or, when relevant, a legally authorized representative (LAR) might sign the ICF on behalf of the study participant 8. Female participants should be: at least 5 years post-menopausal (i.e., persistent amenorrhea 5 years in the absence of an alternative medical cause) or surgically sterile; OR a. Have a negative urine pregnancy test at screening b. Be willing to use a contraceptive method as outlined in inclusion criterion 9 from screening to 30 days after last dose. 9. Male participants who are sexually active with a female partner must agree to the use of an effective method of birth control throughout the study and until 3 months after the last administration of the investigational product. (Note: medically acceptable methods of contraception that may be used by the participant and/or partner include combined oral contraceptive, contraceptive vaginal ring, contraceptive injection, intrauterine device, etonogestrel implant, each supplemented with a condom, as well as sterilization and vasectomy). 10. Female participants who are lactating must agree not to breastfeed during the study and up to 14 days after the intervention. 11. Male participants must agree not to donate sperm for the purpose of reproduction throughout the study and until 3 months after the last administration of the investigational product. 12. For France only: Being affiliated with a European Social Security. Exclusion criteria 1. Not needing or not willing to remain in a healthcare facility during the study 2. Moribund condition (death likely in days) or not expected to survive for >7 days - due to other and non-COVID-19 related conditions 3. Participant on invasive mechanical ventilation via an endotracheal tube, or extracorporeal membrane oxygenation (ECMO), or high-flow Oxygen (delivery of oxygen at a flow of ≥16 L/min.). 4. Participant is not able to take medications by mouth (as capsules or as a powder, mixed in water). 5. Disallowed concomitant medication: Consumption of any herbal products containing 20-hydroxyecdysone and derived from Leuzea carthamoides; Cyanotis vaga or Cyanotis arachnoidea is not allowed (e.g. performance enhancing agents). 6. Any known hypersensitivity to any of the ingredients, or excipients of the study medication, BIO101. 7. Renal disease requiring dialysis, or known renal insufficiency (eGFR≤30 mL/min/1.73 m2, based on Cockcroft & Gault formula). 8. In France only: a. Non-affiliation to compulsory French social security scheme (beneficiary or right-holder). b. Being under tutelage or legal guardianship. Participants will be recruited from approximately 30 clinical centres in Belgium, France, the UK, USA and Brazil. Maximum patients' participation in the study will last 28 days. Follow-up of participants discharged from hospital will be performed through post-intervention phone calls at 14 (± 2) and 60 (± 4) days. INTERVENTION AND COMPARATOR: Two treatment arms will be tested in this study: interventional arm 350 mg b.i.d. of BIO101 (AP 20-hydroxyecdysone) and placebo comparator arm 350 mg b.i.d of placebo. Administration of daily dose is the same throughout the whole treatment period. Participants will receive the study medication while hospitalized for up to 28 days or until a clinical endpoint is reached (i.e., 'negative' or 'positive' event). Participants who are officially discharged from hospital care will no longer receive study medication. MAIN OUTCOMES: Primary study endpoint: The proportion of participants with 'negative' events up to 28 days. 'Negative' events are defined as respiratory deterioration and all-cause mortality. For the purpose of this study, respiratory deterioration will be defined as any of the following: Requiring mechanical ventilation (including cases that will not be intubated due to resource restrictions and triage). Requiring extracorporeal membrane oxygenation (ECMO). Requiring high-flow oxygen defined as delivery of oxygen at a flow of ≥16 L/min. Only if the primary endpoint is significant at the primary final analysis the following Key secondary endpoints will be tested in that order: Proportion of participants with events of respiratory failure at Day 28 Proportion of participants with 'positive' events at Day 28. Proportion of participants with events of all-cause mortality at Day 28 A 'positive' event is defined as the official discharge from hospital care by the department due to improvement in participant condition. Secondary and exploratory endpoints: In addition, a variety of functional measures and biomarkers (including the SpO2 / FiO2 ratio, viral load and markers related to inflammation, muscles, tissue and the RAS / MAS pathways) will also be collected. RANDOMIZATION: Randomization is performed using an IBM clinical development IWRS system during the baseline visit. Block-permuted randomization will be used to assign eligible participants in a 1:1 ratio. In part 1, randomization will be stratified by RAS pathway modulator use (yes/no) and co-morbidities (none vs. 1 and above). In Part 2, randomization will be stratified by centre, gender, RAS pathway modulator use (yes/no), co-morbidities (none vs. 1 and above), receiving Continuous Positive Airway Pressure/Bi-level Positive Airway Pressure (CPAP/BiPAP) at study entry (Yes/No) and suspicion of COVID-19 related myocarditis or pericarditis (present or not). BLINDING (MASKING): Participants, caregivers, and the study team assessing the outcomes are blinded to group assignment. All therapeutic units (TU), BIO101 b.i.d. or placebo b.i.d., cannot be distinguished in compliance with the double-blind process. An independent data-monitoring committee (DMC) will conduct 2 interim analyses. A first one based on the data from part 1 and a second from the data from parts 1 and 2. The first will inform about BIO101 safety, to allow the start of recruitment into part 2 followed by an analysis of the efficacydata, to obtain an indication of activity. The second interim analysis will inform about the sample size that will be required for part 2, in order to achieve adequate statistical power. Numbers to be randomised (sample size) Number of participants randomized: up to 465, in total Part 1: 50 (to obtain the proof of concept in COVID-19 patients). Part 2: 310, potentially increased by 50% (up to 465, based on interim analysis 2) (to confirm the effects of BIO101 observed in part 1). TRIAL STATUS: The current protocol Version is V 10.0, dated on 24.09.2020. The recruitment that started on September 1st 2020 is ongoing and is anticipated to finish for the whole study by March2021. TRIAL REGISTRATION: The trial was registered before trial start in trial registries: EudraCT , No. 2020-001498-63, registered May 18, 2020; and Clinicaltrials.gov, identifier NCT04472728 , registered July 15, 2020. FULL PROTOCOL: The full protocol is attached as an additional file, accessible from the Trials website (Additional file 1). In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.


Assuntos
Tratamento Farmacológico da COVID-19 , Ecdisterona/uso terapêutico , Insuficiência Respiratória/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/fisiopatologia , Progressão da Doença , Método Duplo-Cego , Oxigenação por Membrana Extracorpórea/estatística & dados numéricos , Hospitalização , Humanos , Hipóxia/fisiopatologia , Pessoa de Meia-Idade , Mortalidade , Oxigenoterapia/estatística & dados numéricos , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores de Coronavírus/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistema Renina-Angiotensina , Respiração Artificial/estatística & dados numéricos , Insuficiência Respiratória/fisiopatologia , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/metabolismo , Taquipneia/fisiopatologia , Resultado do Tratamento
3.
Neurosci Res ; 81-82: 21-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24480536

RESUMO

Ecdysterone (EDS), a common derivative of ecdysteroid, has shown its effects on alleviating cognitive impairment and improving the cognition and memory. However, the mechanisms remain unknown. Using temporal global forebrain ischemia and reperfusion-induced brain injury as a model system, we investigated the roles of EDS in improving cognitive impairment in gerbil. Our results demonstrated that intraperitoneal injection of EDS obviously increased the number of surviving neuron cells by Nissl and neuronal nuclei (NeuN) staining. Indeed, the protecting effects of EDS are because of its ability to prevent the apoptosis of neuron cells as evidenced by TUNEL staining and caspase-3 deactivation in the brain of temporal global forebrain ischemia/reperfusion-treated gerbil. Moreover, EDS administration suppressed the ischemia stimulated activity of astrocytes and microglia cells by inhibiting the production of tumor necrosis alpha (TNF-α) in the brain of gerbil. More importantly, these actions of neurons and astrocytes/microglia cells in response to EDS treatment played pivotal roles in ameliorating the cognitive impairment in the ischemia/reperfusion-injured gerbil. In view of these observations, we not only decipher the mechanisms of EDS in reducing the syndrome of ischemia, but also provide novel perspectives to combat ischemic stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Ecdisterona/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Caspase 3/metabolismo , Ecdisterona/farmacologia , Gerbillinae , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
4.
Acta Neurochir Suppl ; 110(Pt 1): 151-5, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21116931

RESUMO

BACKGROUND AND PURPOSE: We reported previously that ecdysterone (EDS) improves neurologic function after experimental stroke. However, the underlying mechanism remained unclear. The present study was conducted to test whether ecdysterone improves neurologic function by enhancing astrocyte activation and angiogenesis after focal cerebral ischemia in rats. METHODS: Focal cerebral ischemia model was conducted by middle cerebral artery occlusion (MCAO). EDS was intraperitoneally injected at 20 mg kg1 daily for 7 days after MCAO. Neurologic recovery was assessed using the neurologic severity scores. Microvessel density and GFAP expression were detected with immunostaining and analyzed quantitatively with image system. RESULTS: Treatment with EDS significantly improved functional recovery, along with increases in density of cerebral microvessels and astrocyte activation. Microvessel density was significantly higher in EDS treated group than in ischemia control group at all time points, and reached a peak on day 14. EDS treated group had substantial increment in GFAP immunoreactive cells, darker staining color, more and longer nerve processes, higher GFAP expression and area of immunoreactive cells at each time point. CONCLUSION: Our data suggest that EDS treatment enhanced angiogenesis and astrocyte activation which could contribute to functional recovery.


Assuntos
Astrócitos/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Ecdisterona/uso terapêutico , Neovascularização Fisiológica/efeitos dos fármacos , Análise de Variância , Animais , Modelos Animais de Doenças , Ecdisterona/farmacologia , Fator VIII/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Microvasos/efeitos dos fármacos , Microvasos/patologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Índice de Gravidade de Doença , Fatores de Tempo
5.
J Alzheimers Dis ; 22(1): 107-17, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20847437

RESUMO

Amyloid-ß (Aß) plays a pivotal role in Alzheimer's disease (AD) pathogenesis and in toxic mechanisms such as oxidative stress, mitochondrial dysfunction, calcium turbulence, and apoptosis induction. Therefore, interfering with Aß aggregation has long been one of the most promising strategies for AD treatment. Ecdysterones (ECRs) are steroidal hormones in insects and terrestrial plants that have high structural diversity and multiple beneficial pharmacological activities. Here, we studied the effects of six ECRs on Aß aggregation and cytotoxicity. Two ECRs with an acetoxyl group at the 2 or 3 position and saturated chains as side groups showed apparent promotion of Aß42 fibrilization, resulting in less Aß42 oligomers in the samples. Another three with unsaturated side chains clearly inhibited Aß aggregation and disaggregated preformed fibrils, but increased the Aß42 oligomer levels. Nevertheless, our MTT results showed that all ECRs tested inhibited Aß42-induced cytotoxicity. This protective activity may be partly attributable to ECR-mediated amelioration of A&beta42-induced release of reactive oxygen species. Taken together, our findings suggest that ECRs, a series of natural compounds in many plants and insects, have therapeutic potential in AD and that the deduced structure-activity relationships may be beneficial in drug design for the treatment of AD and other amyloidoses.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Ecdisterona/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/toxicidade , Multimerização Proteica/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Linhagem Celular Tumoral , Ecdisterona/química , Ecdisterona/uso terapêutico , Humanos , Fragmentos de Peptídeos/metabolismo , Dobramento de Proteína/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade
6.
Phytomedicine ; 17(11): 884-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20554186

RESUMO

Estrogens exert beneficial effects in the bone. Their chronic use however bares several risks. Therefore intensive search for non-estrogenic, bone protective compounds is going on. We observed that an extract of Tinospora cordifolia has antiosteoporotic effects and identified 20-OH-Ecdysone (beta-Ecdysone=Ecd) as a possible candidate for this action. Ovariectomized (ovx) rats were treated orally over 3 months with no Ecd (control) or 18, 57 or 121 mg Ecd/day/animal. Estradiol-17beta benzoate (E2) 159 microg/day/animal) fed animals served as positive controls. Bone mineral density (BMD) of tibia was measured by quantitative computer tomography, serum Osteocalcin and CrossLaps were measured in a ligand binding assay. Utilizing an estrogen receptor (ER) containing cytosolic extract of porcine uteri the capability of Ecd to bind to ER was tested. Ecd did not bind to ER. BMD was reduced by more than 50% in the control. In the Ecd animals BMD was dose dependently higher. Serum CrossLaps was lower in the Ecd and E2 group while serum Osteocalcin levels were decreased in the E2 but increased in the Ecd fed animals. Ecd has an antiosteoporotic effect which does not involve activation of ER.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Densidade Óssea/efeitos dos fármacos , Ecdisterona/farmacologia , Osteoporose/prevenção & controle , Extratos Vegetais/farmacologia , Receptores de Estrogênio/metabolismo , Tinospora/química , Animais , Conservadores da Densidade Óssea/uso terapêutico , Colágeno/sangue , Relação Dose-Resposta a Droga , Ecdisterona/uso terapêutico , Feminino , Osteocalcina/sangue , Osteoporose/sangue , Ovariectomia , Fragmentos de Peptídeos/sangue , Fitoterapia , Extratos Vegetais/uso terapêutico , Ratos , Ratos Sprague-Dawley , Suínos , Útero/efeitos dos fármacos , Útero/patologia
7.
Phytomedicine ; 17(5): 350-5, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20171072

RESUMO

Ecdysteroids are steroids found in invertebrates and plants. In mammals they have protein anabolic effects. We have recently published antiosteoporotic effects of Tinospora cordifolia (TC) extract and the search for the possible active ingredients yielded the presence of beta-Ecdysone (Ecd). Therefore, we investigated the effects of pure Ecd in ovariectomized rats on morphological changes in joint, epiphyseal cartilage and trabecular tissue. Following ovariectomy rats were fed for 1 month with Ecd containing food at a dose of 52.8 mg/day/animal. Positive and negative control animals received 17-beta Estradiol (E(2), 132 microg/day/animal) and soy free (sf) food respectively. At sacrifice, specimens consisting of upper tibiae-lower femurs and knee joint were harvested and processed for histomorphometry. The parameters measured included thickness of the joint cartilage, thickness of the whole epiphyseal growth plate and its three zones. Furthermore, the percentage of trabecular bone in the metaphysis region of tibiae was quantified. Ecd and E(2) induced a significant increase in the thickness of joint cartilage. The whole epiphyseal growth plate and its proliferative and hypertrophic zones were also increased by Ecd whereas E(2) reduced their size. The percentage of trabecular area in the metaphysis of tibia was significantly increased in Ecd and E(2) treated animals. Results provide a plausible explanation for the antiosteoporotic effects of TC. Hence, TC as well as other Ecd producing plants or pure Ecd may be of value in the prevention and treatment of osteoporosis and osteoarthritis which is of increasing importance due to aging and obesity among individuals.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Osso e Ossos/efeitos dos fármacos , Ecdisterona/farmacologia , Lâmina de Crescimento/efeitos dos fármacos , Articulação do Joelho/efeitos dos fármacos , Extratos Vegetais/farmacologia , Tinospora/química , Animais , Densidade Óssea/efeitos dos fármacos , Conservadores da Densidade Óssea/uso terapêutico , Dieta , Quimioterapia Combinada , Ecdisterona/uso terapêutico , Estradiol/farmacologia , Estradiol/uso terapêutico , Feminino , Fêmur/efeitos dos fármacos , Osteoartrite do Joelho/tratamento farmacológico , Osteoporose/tratamento farmacológico , Osteoporose/prevenção & controle , Ovariectomia , Fitoterapia , Extratos Vegetais/uso terapêutico , Ratos , Ratos Wistar , Alimentos de Soja , Tíbia/efeitos dos fármacos
8.
Biol Pharm Bull ; 31(12): 2245-9, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19043207

RESUMO

The effect on bone tissue of beta-ecdysterone, a type of ecdysteroid found in many plants, has not been previously investigated. In this study, we found that beta-ecdysterone treatment significantly induced alkaline phosphatase (ALP) activity in mesenchymal stem cells in a dose-dependent manner. Real-time polymerase chain reaction (PCR) showed that Runx2, osteocalcin, and type I collagen expression also increased. ICI182780, a specific estrogen receptor antagonist, inhibited the upregulation of ALP activity. Moreover, beta-ecdysterone promoted estrogen receptor (ER) reporter gene activity; however, ICI182780 reversed its effect, suggesting that beta-ecdysterone has stimulatory effects on osteogenic differentiation via the ER. Furthermore, beta-ecdysterone alleviated osteoporosis symptoms in a mouse model without obvious side effects. Therefore beta-ecdysterone may be a promising candidate drug for the treatment of osteoporosis.


Assuntos
Conservadores da Densidade Óssea , Ecdisterona/farmacologia , Ecdisterona/uso terapêutico , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteoporose/prevenção & controle , Fosfatase Alcalina/metabolismo , Animais , Compostos Azo , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/química , Osso e Ossos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Corantes , Genes Reporter/efeitos dos fármacos , Indicadores e Reagentes , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Osteoporose/induzido quimicamente , Osteoporose/patologia , Receptores de Estrogênio/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sais de Tetrazólio , Tiazóis , Transfecção , Tretinoína
9.
Ukr Biokhim Zh (1999) ; 80(1): 46-51, 2008.
Artigo em Ucraniano | MEDLINE | ID: mdl-18710026

RESUMO

Chronic hyperglycaemia (60 days) which developed after streptozotocine (STZ) administration (5 mg/100 g) in rats was accompanied with development of severe endothelial dysfunction as well as with disturbed non-haem iron metabolism. It was established by EPR spectroscopy method that STZ administration reduced transferrin levels in the blood as well as pools of iron associated with blood transferrin and with ferritin in the heart and aorta of rats with hyperglycaemia. Chronic ecdysterone administration (100 ng/100 g, 60 days) protects hyperglycaemia development by preventing of non-haem iron metabolism disturbance. These data suppose participation of non-haem iron in mechanisms of ecdysterone protection of streptozotocine-induced hyperglycaemia and ischemia.


Assuntos
Ecdisterona/uso terapêutico , Hiperglicemia/prevenção & controle , Ferro/metabolismo , Ferroproteínas não Heme/metabolismo , Substâncias Protetoras/uso terapêutico , Animais , Glicemia/análise , Ecdisterona/administração & dosagem , Ecdisterona/farmacologia , Espectroscopia de Ressonância de Spin Eletrônica , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Hiperglicemia/sangue , Hiperglicemia/metabolismo , Ferro/sangue , Masculino , Substâncias Protetoras/administração & dosagem , Substâncias Protetoras/farmacologia , Ratos , Ratos Endogâmicos WKY , Estreptozocina , Transferrina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA