Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 14(1): 69, 2023 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-37024935

RESUMO

BACKGROUND: There remains much interest in improving cryopreservation techniques for advanced therapy medicinal products (ATMPs). Recently, human platelet lysate (hPL) has emerged as a promising candidate to replace fetal bovine serum (FBS) as a xeno-free culture supplement for the expansion of human cell therapy products. Whether hPL can also substitute for FBS in cryopreservation procedures remains poorly studied. Here, we evaluated several cryoprotective formulations based on a proprietary hPL for the cryopreservation of bioengineered tissues and cell therapy products. METHODS: We tested different xenogeneic-free, pathogen-inactivated hPL (ihPL)- and non-inactivated-based formulations for cryopreserving bioengineered tissue (cellularized nanostructured fibrin agarose hydrogels (NFAHs)) and common cell therapy products including bone marrow-derived mesenchymal stromal cells (BM-MSCs), human dermal fibroblasts (FBs) and neural stem cells (NSCs). To assess the tissue and cellular properties post-thaw of NFAHs, we analyzed their cell viability, identity and structural and biomechanical properties. Also, we evaluated cell viability, recovery and identity post-thaw in cryopreserved cells. Further properties like immunomodulation, apoptosis and cell proliferation were assessed in certain cell types. Additionally, we examined the stability of the formulated solutions. The formulations are under a bidding process with MD Bioproducts (Zurich, Switzerland) and are proprietary. RESULTS: Amongst the tissue-specific solutions, Ti5 (low-DMSO and ihPL-based) preserved the viability and the phenotype of embedded cells in NFAHs and preserved the matrix integrity and biomechanical properties similar to those of the standard cryopreservation solution (70% DMEM + 20% FBS + 10% DMSO). All solutions were stable at - 20 °C for at least 3 months. Regarding cell-specific solutions, CeA maintained the viability of all cell types > 80%, preserved the immunomodulatory properties of BM-MSCs and promoted good recovery post-thaw. Besides, both tested solutions were stable at - 20 °C for 18 months. Finally, we established that there is a 3-h window in which thawed NFAHs and FBs maintain optimum viability immersed in the formulated solutions and at least 2 h for BM-MSCs. CONCLUSIONS: Our results show that pathogen-inactivated solutions Ti5 allocated for bioengineered tissues and CeA allocated for cells are efficient and safe candidates to cryopreserve ATMPs and offer a xenogeneic-free and low-DMSO alternative to commercially available cryoprotective solutions.


Assuntos
Técnicas de Cultura de Células , Dimetil Sulfóxido , Humanos , Técnicas de Cultura de Células/métodos , Plaquetas/química , Células Cultivadas , Proliferação de Células/genética , Criopreservação/métodos , Terapia Baseada em Transplante de Células e Tecidos , Diferenciação Celular/genética
2.
Lancet Neurol ; 22(2): 137-146, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36681446

RESUMO

BACKGROUND: Pilot clinical trials have shown the safety of intra-arterial bone marrow mononuclear cells (BMMNCs) in stroke. However, the efficacy of different doses of intra-arterial BMMNCs in patients with acute stroke has not been tested in a randomised clinical trial. We aimed to show safety and efficacy of two different doses of autologous intra-arterial BMMNC transplantation in patients with acute stroke. METHODS: The IBIS trial was a multicentre phase 2, randomised, controlled, investigator-initiated, assessor-blinded, clinical trial, in four stroke centres in Spain. We included patients (aged 18-80 years) with a non-lacunar, middle cerebral artery ischaemic stroke within 1-7 days from stroke onset and with a National Institutes of Health Stroke Scale score of 6-20. We randomly assigned patients (2:1:1) with a computer-generated randomisation sequence to standard of care (control group) or intra-arterial injection of autologous BMMNCs at one of two different doses (2 × 106 BMMNCs/kg or 5 × 106 BMMNCs/kg). The primary efficacy outcome was the proportion of patients with modified Rankin Scale scores of 0-2 at 180 days in the intention-to-treat population, comparing each BMMNC dose group and the pooled BMMNC group versus the control group. The primary safety endpoint was the proportion of serious adverse events. This trial was registered at ClinicalTrials.gov, NCT02178657 and is completed. FINDINGS: Between April 1, 2015, and May 20, 2021, we assessed 114 patients for eligibility. We randomly assigned 77 (68%) patients: 38 (49%) to the control group, 20 (26%) to the low-dose BMMNC group, and 19 (25%) the high-dose BMMNC group. The mean age of participants was 62·4 years (SD 12·7), 46 (60%) were men, 31 (40%) were women, all were White, and 63 (82%) received thrombectomy. The median NIHSS score before randomisation was 12 (IQR 9-15), with intra-arterial BMMNC injection done a median of 6 days (4-7) after stroke onset. The primary efficacy outcome occurred in 14 (39%) patients in the control group versus ten (50%) in the low-dose group (adjusted odds ratio 2·08 [95% CI 0·55-7·85]; p=0·28), eight (44%) in the high-dose group (1·89 [0·52-6·96]; p=0·33), and 18 (47%) in the pooled BMMNC group (2·22 [0·72-6·85]; p=0·16). We found no differences in the proportion of patients who had adverse events or dose-related events, but two patients had a groin haematoma after cell injection in the low-dose BMMNC group. INTERPRETATION: Intra-arterial BMMNCs were safe in patients with acute ischaemic stroke, but we found no significant improvement at 180 days on the mRS. Further clinical trials are warranted to investigate whether improvements might be possible at different timepoints. FUNDING: Instituto de Salud Carlos III co-funded by the European Regional Development Fund/European Social Fund, Mutua Madrileña, and the Regional Ministry of Health of Andalusia.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Masculino , Humanos , Feminino , Pessoa de Meia-Idade , Acidente Vascular Cerebral/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Espanha , Medula Óssea , Resultado do Tratamento , Transplante de Células
3.
Front Bioeng Biotechnol ; 10: 908250, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36082161

RESUMO

Fibrin is widely used for tissue engineering applications. The use of blood derivatives, however, carries a high risk of transmission of infectious agents, necessitating the application of pathogen reduction technology (PRT). The impact of this process on the structural and biomechanical properties of the final products is unknown. We used normal plasma (PLc) and plasma inactivated by riboflavin and ultraviolet light exposure (PLi) to manufacture nanostructured cellularized fibrin-agarose hydrogels (NFAHs), and then compared their structural and biomechanical properties. We also measured functional protein C, prothrombin time (PT), activated partial thromboplastin time (APTT), thrombin time (TT) and coagulation factors [fibrinogen, Factor (F) V, FVIII, FX, FXI, FXIII] in plasma samples before and after inactivation. The use of PLi to manufacture cellularized NFAHs increased the interfibrillar spacing and modified their biomechanical properties as compared with cellularized NFAH manufactured with PLc. PLi was also associated with a significant reduction in functional protein C, FV, FX, and FXI, and an increase in the international normalized ratio (derived from the PT), APTT, and TT. Our findings demonstrate that the use of PRT for fibrin-agarose bioartificial tissue manufacturing does not adequately preserve the structural and biomechanical properties of the product. Further investigations into PRT-induced changes are warranted to determine the applications of NFAH manufactured with inactivated plasma as a medicinal product.

4.
Platelets ; 33(1): 98-109, 2022 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-33393414

RESUMO

Recent years have witnessed the introduction of ex vivo expanded dermal fibroblasts for several cell therapy and tissue-engineering applications, including the treatment of facial scars and burns, representing a promising cell type for regenerative medicine. We tested different in-house produced human platelet lysate (HPL) solutions against fetal bovine serum as supplements for in vitro fibroblast expansion by comparing cell yield, molecular marker expression, extracellular matrix (ECM) generation, genomic stability and global gene expression. Our in-house produced HPL supported fibroblast growth at levels similar to those for FBS and commercial HPL products and was superior to AB human serum. Cells grown in HPL maintained a fibroblast phenotype (VIM+, CD44+, CD13+, CD90+), ECM generation capacity (FN+, COL1+) and a normal karyotype, although gene expression profiling revealed changes related to cell metabolism, adhesion and cellular senescence. The HPL manufacturing process was validated within a GMP compliant system and the solution was stable at -80ºC and -20ºC for 2 years. Dermal fibroblasts expanded in vitro with HPL maintain a normal karyotype and expression of fibroblast markers, with only minor changes in their global gene expression profile. Our in-house produced GMP-HPL is an efficient, safe and economical cell culture supplement that can help increase the healthcare activity of blood transfusion centers through the re-use of transfusional plasma and platelets approaching their expiration date. Currently, our HPL solution is approved by the Spanish Agency of Medicines and Medical Devices and is being used in the manufacture of cell therapy products.Abbreviations: AB plasma: plasma group AB; ABHS: AB Human Serum; ABHS+GF: AB Human Serum supplemented with growth factors; ANOVA: Analysis of variance; ATMPs: Advanced Therapies for Medicinal Products; CPE: cytopathic effect; DEGs: Differentially expressed genes; DMEM: Dulbecco's modified Eagle's Medium; ECM: Extracellular matrix; ELISA: enzyme-linked immunosorbent assay; FBS: Fetal bovine serum; FDR: False discovery rate; FGF: Fibroblast growth factor; GMP: Good manufacturing practice; HPL: Human platelet lysate; HPL-CM: commercial human platelet lysate; MSCs: mesenchymal stem cells; NEAA: non-essential amino acids; P/S: penicillin/streptomycin; PBS: phosphate buffered saline; PC: leukodepleted platelet concentrate; PCR: polymerase chain reaction; PDGF: Platelet-derived growth factor; PDGFRA: Platelet-derived growth factor receptor alpha; qPCR: quantitative polymerase chain reaction; RNA: Ribonucleic acid; RT: Room temperature; TAC: Transcriptome analysis console; TGF-ß: Transforming growth factor beta.


Assuntos
Plaquetas/metabolismo , Fibroblastos/metabolismo , Animais , Bovinos , Feto , Humanos
5.
J Tissue Eng Regen Med ; 13(4): 664-673, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30793853

RESUMO

Blood loss remains a major concern during surgery and can increase the morbidity of the intervention. The use of topical haemostatic agents to overcome this issue therefore becomes necessary. Fibrin sealants are promising haemostatic agents due to their capacity to promote coagulation, but their effectiveness and applicability need to be improved. We have compared the haemostatic efficacy of a novel nanostructured fibrin-agarose hydrogel patch, with (c-NFAH) or without cells (a-NFAH), against two commercially available haemostatic agents in a rat model of hepatic resection. Hepatic resections were performed by making short or long incisions (mild or severe model, respectively), and haemostatic agents were applied to evaluate time to haemostasis, presence of haematoma, post-operative adhesions to adjacent tissues, and inflammation factors. We found a significantly higher haemostatic success rate (time to haemostasis) with a-NFAH than with other commercial haemostatic agents. Furthermore, other relevant outcomes investigated were also improved in the a-NFAH group, including no presence of haematoma, lower adhesions, and lower grades of haemorrhage, inflammation, and necrosis in histological analysis. Overall, these findings identify a-NFAH as a promising haemostatic agent in liver resection and likely in a range of surgical procedures.


Assuntos
Fibrina/farmacologia , Hemostáticos/farmacologia , Hidrogéis/farmacologia , Nanoestruturas/química , Sefarose/farmacologia , Animais , Hemorragia/patologia , Inflamação/patologia , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Necrose , Ratos Wistar
6.
Age (Dordr) ; 34(2): 389-403, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21404051

RESUMO

Coenzyme Q10 (CoQ) is a powerful antioxidant that reduces oxidative stress. We explored whether the quality of dietary fat alters postprandial oxidative DNA damage and whether supplementation with CoQ improves antioxidant capacity by modifying the activation/stabilization of p53 in elderly subjects. In this crossover study, 20 subjects were randomly assigned to receive three isocaloric diets during 4 weeks each: (1) Mediterranean diet (Med diet), (2) Mediterranean diet supplemented with CoQ (Med+CoQ diet), and (3) saturated fatty acid-rich diet (SFA diet). Levels of mRNAs were determined for p53, p21, p53R2, and mdm2. Protein levels of p53, phosphorylated p53 (Ser20), and monoubiquitinated p53 were also measured, both in cytoplasm and nucleus. The extent of DNA damage was measured as plasma 8-OHdG. SFA diet displayed higher postprandial 8-OHdG concentrations, p53 mRNA and monoubiquitinated p53, and lower postprandial Mdm2 mRNA levels compared with Med and Med+CoQ diets (p < 0.05). Moreover, Med+CoQ diet induced a postprandial decrease of cytoplasmatic p53, nuclear p-p53 (Ser20), and nuclear and cytoplasmatic monoubiquitinated p53 protein (p < 0.05). In conclusion, Med+CoQ diet improves oxidative DNA damage in elderly subjects and reduces processes of cellular oxidation. Our results suggest a starting point for the prevention of oxidative processes associated with aging.


Assuntos
Envelhecimento/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Dieta Mediterrânea , Genes p53/genética , Estresse Oxidativo/fisiologia , Período Pós-Prandial/efeitos dos fármacos , Ubiquinona/análogos & derivados , Idoso , Envelhecimento/metabolismo , Western Blotting , Estudos Cross-Over , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Regulação da Expressão Gênica , Genes p53/efeitos dos fármacos , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases , Masculino , Oxirredução , Período Pós-Prandial/genética , Reação em Cadeia da Polimerase em Tempo Real , Ubiquinona/administração & dosagem , Ubiquinona/farmacocinética , Vitaminas/administração & dosagem , Vitaminas/farmacocinética
7.
Clin Dev Immunol ; 2012: 974648, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21941583

RESUMO

Autoimmune diseases (AIDs) have been associated with accelerated atherosclerosis (AT) leading to increased cardio- and cerebrovascular disease risk. Traditional risk factors, as well as systemic inflammation mediators, including cytokines, chemokines, proteases, autoantibodies, adhesion receptors, and others, have been implicated in the development of these vascular pathologies. Yet, the characteristics of vasculopathies may significantly differ depending on the underlying disease. In recent years, many new genes and signalling pathways involved in autoimmunity with often overlapping patterns between different disease entities have been further detected. Epigenetics, the control of gene packaging and expression independent of alterations in the DNA sequence, is providing new directions linking genetics and environmental factors. Epigenetic regulatory mechanisms comprise DNA methylation, histone modifications, and microRNA activity, all of which act upon gene and protein expression levels. Recent findings have contributed to our understanding of how epigenetic modifications could influence AID development, not only showing differences between AID patients and healthy controls, but also showing how one disease differs from another and even how the expression of key proteins involved in the development of each disease is regulated.


Assuntos
Doenças Autoimunes/epidemiologia , Doenças Cardiovasculares/epidemiologia , Epigênese Genética , Acetilação , Doenças Autoimunes/complicações , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/imunologia , Metilação de DNA , Previsões , Inibidores de Histona Desacetilases/uso terapêutico , Histonas/metabolismo , Humanos , Metilação , MicroRNAs/fisiologia , Processamento de Proteína Pós-Traducional , Doenças Reumáticas/complicações , Doenças Reumáticas/epidemiologia , Doenças Reumáticas/genética , Doenças Reumáticas/imunologia , Risco
8.
Age (Dordr) ; 34(2): 341-58, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21472381

RESUMO

Fatty acids and other components of the diet may modulate, among others, mechanisms involved in homeostasis, aging, and age-related diseases. Using a proteomic approach, we have studied how dietary oil affected plasma proteins in young (6 months) or old (24 months) rats fed lifelong with two experimental diets enriched in either sunflower or virgin olive oil. After the depletion of the most abundant proteins, levels of less abundant proteins were studied using two-dimensional electrophoresis and mass spectrometry. Our results showed that compared with the sunflower oil diet, the virgin olive oil diet induced significant decreases of plasma levels of acute phase proteins such as inter-alpha inhibitor H4P heavy chain (at 6 months), hemopexin precursor (at 6 and 24 months), preprohaptoglobin precursor (at 6 and 24 months), and α-2-HS glycoprotein (at 6 and 24 months); antioxidant proteins such as type II peroxiredoxin (at 24 months); proteins related with coagulation such as fibrinogen γ-chain precursor (at 24 months), T-kininogen 1 precursor (at 6 and 24 months), and apolipoprotein H (at 6 and 24 months); or with lipid metabolism and transport such as apolipoprotein E (at 6 and 24 months) and apolipoprotein A-IV (at 24 months). The same diet increased the levels of apolipoprotein A-1 (at 6 and 24 months), diminishing in general the changes that occurred with age. Our unbiased analysis reinforces the beneficial role of a diet rich in virgin olive oil compared with a diet rich in sunflower oil, modulating inflammation, homeostasis, oxidative stress, and cardiovascular risk during aging.


Assuntos
Envelhecimento/sangue , Gorduras Insaturadas na Dieta/farmacologia , Óleos de Plantas/farmacologia , Proteoma/metabolismo , Envelhecimento/efeitos dos fármacos , alfa-Globulinas/metabolismo , Animais , Antioxidantes/metabolismo , Western Blotting , Eletroforese em Gel Bidimensional , Haptoglobinas , Hemopexina/metabolismo , Masculino , Espectrometria de Massas , Azeite de Oliva , Precursores de Proteínas/sangue , Proteoma/efeitos dos fármacos , Ratos , Ratos Wistar , alfa-2-Glicoproteína-HS/metabolismo
9.
Clín. investig. arterioscler. (Ed. impr.) ; 23(6): 262-268, nov.-dic. 2011. tab
Artigo em Espanhol | IBECS | ID: ibc-96045

RESUMO

Introducción Los estudios realizados hasta la fecha, sobre los mecanismos de acción de los compuestos fenólicos, indican que sus efectos podrían deberse no sólo a la eliminación de radicales libres, sino que modularían los procesos de señalización celular o actuarían por sí mismos como moléculas señal. Métodos Se administraron 2 desayunos basados en aceite de oliva, uno de ellos con alto contenido en compuestos fenólicos (398ppm) y el otro con bajo contenido (70ppm), a 20 pacientes con síndrome metabólico, en un diseño aleatorio y cruzado. Se analizó la expresión génica postprandial en células mononucleares de sangre periférica, mediante microarrays. Resultados Hemos observado que el desayuno que incluyó un aceite rico en compuestos fenólicos reprimió la expresión de 18 genes relacionados directamente con procesos de señalización, entre ellos 12 factores de transcripción implicados en proliferación y crecimiento celular. Conclusiones Nuestros resultados sugieren que el consumo de un desayuno con aceite de oliva virgen, rico en compuestos fenólicos, reduce los procesos de proliferación celular en células mononucleares, lo que podría ser un mecanismo de protección del desarrollo de arteriosclerosis (AU)


Introduction It has been speculated that the potential beneficial effects of olive oil could be due to modulation of genes involved in proliferative, antioxidant and inflammatory pathways. Research on the acting and mechanisms of phenolic compounds indicates that the beneficial effects associated with these compounds may not only be due to the elimination of free radicals, but could also modulate cell signalling processes, or could themselves act as signaling molecules. Methods Two virgin olive oil-based breakfasts with high (398ppm) and low (70ppm) content of phenolic compounds were administered to 20 patients with metabolic syndrome following a double-blinded random crossover design. Postprandial gene expression in peripheral blood mononuclear cells was analyzed at 4hours by gene expression microarrays. Results We observed that the phenolic compounds of olive oil repressed the expression of 18 genes directly related to signalling processes, including 12 transcription factors involved in proliferation and cell growth. Conclusions Our results suggest that consumption of olive oil rich in phenolic compounds reduces the processes of cell proliferation in mononuclear cells in the postprandial state, which could reduce development of atherosclerosis (AU)


Assuntos
Humanos , Compostos Fenólicos/análise , Gorduras Vegetais , Síndrome Metabólica/metabolismo , Aterosclerose/fisiopatologia , Elementos Reguladores de Transcrição , Proliferação de Células , Radicais Livres , Antioxidantes/farmacocinética
10.
Age (Dordr) ; 33(4): 579-90, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21170684

RESUMO

Postprandial oxidative stress is characterized by an increased susceptibility of the organism towards oxidative damage after consumption of a meal rich in lipids and/or carbohydrates. We have investigated whether the quality of dietary fat alters postprandial cellular oxidative stress and whether the supplementation with coenzyme Q(10) (CoQ) lowers postprandial oxidative stress in an elderly population. In this randomized crossover study, 20 participants were assigned to receive three isocaloric diets for periods of 4 week each: (1) Mediterranean diet supplemented with CoQ (Med+CoQ diet), (2) Mediterranean diet (Med diet), and (3) saturated fatty acid-rich diet (SFA diet). After a 12-h fast, the volunteers consumed a breakfast with a fat composition similar to that consumed in each of the diets. CoQ, lipid peroxides (LPO), oxidized low-density lipoprotein (oxLDL), protein carbonyl (PC), total nitrite, nitrotyrosine plasma levels, catalase, superoxide dismutase (SOD), and glutathione peroxidase (GPx) activities and ischemic reactive hyperaemia (IRH) were determined. Med diet produced a lower postprandial GPx activity and a lower decrease in total nitrite level compared to the SFA diet. Med and Med+CoQ diets induced a higher postprandial increase in IRH and a lower postprandial LPO, oxLDL, and nitrotyrosine plasma levels than the SFA diet. Moreover, the Med+CoQ diet produced a lower postprandial decrease in total nitrite and a greater decrease in PC levels compared to the other two diets and lower SOD, CAT, and GPx activities than the SFA diet.In conclusion, Med diet reduces postprandial oxidative stress by reducing processes of cellular oxidation and increases the action of the antioxidant system in elderly persons and the administration of CoQ further improves this redox balance.


Assuntos
Dieta Mediterrânea , Suplementos Nutricionais , Estresse Oxidativo , Período Pós-Prandial , Ubiquinona/análogos & derivados , Vitaminas/administração & dosagem , Idoso , Apolipoproteínas/sangue , Glicemia/análise , Estudos Cross-Over , Gorduras na Dieta/administração & dosagem , Endotélio Vascular/fisiologia , Feminino , Humanos , Insulina/sangue , Fluxometria por Laser-Doppler , Lipídeos/sangue , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ubiquinona/administração & dosagem
11.
BMC Genomics ; 11: 253, 2010 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-20406432

RESUMO

BACKGROUND: Previous studies have shown that acute intake of high-phenol virgin olive oil reduces pro-inflammatory, pro-oxidant and pro-thrombotic markers compared with low phenols virgin olive oil, but it still remains unclear whether effects attributed to its phenolic fraction are exerted at transcriptional level in vivo. To achieve this goal, we aimed at identifying expression changes in genes which could be mediated by virgin olive oil phenol compounds in the human. RESULTS: Postprandial gene expression microarray analysis was performed on peripheral blood mononuclear cells during postprandial period. Two virgin olive oil-based breakfasts with high (398 ppm) and low (70 ppm) content of phenolic compounds were administered to 20 patients suffering from metabolic syndrome following a double-blinded, randomized, crossover design. To eliminate the potential effect that might exist in their usual dietary habits, all subjects followed a similar low-fat, carbohydrate rich diet during the study period. Microarray analysis identified 98 differentially expressed genes (79 underexpressed and 19 overexpressed) when comparing the intake of phenol-rich olive oil with low-phenol olive oil. Many of these genes seem linked to obesity, dyslipemia and type 2 diabetes mellitus. Among these, several genes seem involved in inflammatory processes mediated by transcription factor NF-kappaB, activator protein-1 transcription factor complex AP-1, cytokines, mitogen-activated protein kinases MAPKs or arachidonic acid pathways. CONCLUSION: This study shows that intake of virgin olive oil based breakfast, which is rich in phenol compounds is able to repress in vivo expression of several pro-inflammatory genes, thereby switching activity of peripheral blood mononuclear cells to a less deleterious inflammatory profile. These results provide at least a partial molecular basis for reduced risk of cardiovascular disease observed in Mediterranean countries, where virgin olive oil represents a main source of dietary fat. Admittedly, other lifestyle factors are also likely to contribute to lowered risk of cardiovascular disease in this region.


Assuntos
Gorduras na Dieta/uso terapêutico , Perfilação da Expressão Gênica , Leucócitos Mononucleares/metabolismo , Síndrome Metabólica/dietoterapia , Síndrome Metabólica/genética , Óleos de Plantas/uso terapêutico , Redes Reguladoras de Genes , Humanos , Leucócitos Mononucleares/química , Redes e Vias Metabólicas , Síndrome Metabólica/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Azeite de Oliva , Fenóis/análise , Óleos de Plantas/química , Período Pós-Prandial
12.
Expert Opin Investig Drugs ; 19(4): 535-54, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20367194

RESUMO

IMPORTANCE OF THE FIELD: Coenzyme Q(10) (CoQ(10)) is found in blood and in all organs. CoQ(10) deficiencies are due to autosomal recessive mutations, ageing-related oxidative stress and carcinogenesis processes, and also statin treatment. Many neurodegenerative disorders, diabetes, cancer and muscular and cardiovascular diseases have been associated with low CoQ(10) levels, as well as different ataxias and encephalomyopathies. AREAS COVERED IN THIS REVIEW: We review the efficacy of a variety of commercial formulations which have been developed to solubilise CoQ(10) and promote its better absorption in vivo, and its use in the therapy of pathologies associated with low CoQ(10) levels, with emphasis in the results of the clinical trials. Also, we review the use of its analogues idebenone and MitoQ. WHAT THE READER WILL GAIN: This review covers the most relevant aspects related with the therapeutic use of CoQ(10), including existing formulations and their effects on its bioavailability. TAKE HOME MESSAGE: CoQ(10) does not cause serious adverse effects in humans and new formulations have been developed that increase CoQ(10) absorption. Oral CoQ(10) is a viable antioxidant strategy in many diseases, providing a significant to mild symptomatic benefit. Idebenone and MitoQ are promising substitutive CoQ(10)-related drugs which are well tolerated and safe.


Assuntos
Química Farmacêutica , Ubiquinona/química , Ubiquinona/uso terapêutico , Animais , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/genética , Química Farmacêutica/métodos , Humanos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/genética , Ubiquinona/deficiência
13.
Chem Biol Interact ; 181(1): 95-106, 2009 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-19523936

RESUMO

D-Galactosamine (D-GalN) induces reactive oxygen species (ROS) generation and cell death in cultured hepatocytes. The aim of the study was to evaluate the cytoprotective properties of N-acetylcysteine (NAC), coenzyme Q(10) (Q(10)) and the superoxide dismutase (SOD) mimetic against the mitochondrial dysfunction and cell death in D-GalN-treated hepatocytes. Hepatocytes were isolated from liver resections. NAC (0.5 mM), Q(10) (30 microM) or MnTBAP (Mn(III)tetrakis(4-benzoic acid) porphyrin chloride (1mg/mL) were co-administered with D-GalN (40 mM) in hepatocytes. Cell death, oxidative stress, mitochondrial transmembrane potential (MTP), ATP, mitochondrial oxidized/reduced glutathione (GSH) and Q(10) ratios, electronic transport chain (ETC) activity, and nuclear- and mitochondria-encoded expression of complex I subunits were determined in hepatocytes. d-GalN induced a transient increase of mitochondrial hyperpolarization and oxidative stress, followed by an increase of oxidized/reduced GSH and Q(10) ratios, mitochondrial dysfunction and cell death in hepatocytes. The cytoprotective properties of NAC supplementation were related to a reduction of ROS generation and oxidized/reduced GSH and Q(10) ratios, and a recovery of mitochondrial complexes I+III and II+III activities and cellular ATP content. The co-administration of Q(10) or MnTBAP recovered oxidized/reduced GSH ratio, and reduced ROS generation, ETC dysfunction and cell death induced by D-GalN. The cytoprotective properties of studied antioxidants were related to an increase of the protein expression of nuclear- and mitochondrial-encoded subunits of complex I. In conclusion, the co-administration of NAC, Q(10) and MnTBAP enhanced the expression of complex I subunits, and reduced ROS production, oxidized/reduced GSH ratio, mitochondrial dysfunction and cell death induced by D-GalN in cultured hepatocytes.


Assuntos
Acetilcisteína/farmacologia , Galactosamina/farmacologia , Hepatócitos/efeitos dos fármacos , Mitocôndrias Hepáticas/efeitos dos fármacos , Superóxido Dismutase/farmacologia , Ubiquinona/análogos & derivados , Trifosfato de Adenosina/metabolismo , Caspase 3/metabolismo , Células Cultivadas , Feminino , Glutationa/metabolismo , Hepatócitos/patologia , Humanos , L-Lactato Desidrogenase/metabolismo , Masculino , Pessoa de Meia-Idade , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Hepáticas/fisiologia , Mimetismo Molecular , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Ubiquinona/farmacologia
14.
Exp Gerontol ; 42(8): 798-806, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17587521

RESUMO

Dietary coenzyme Q(10) prolongs life span of rats fed on a PUFAn-6-enriched diet. Our aim was to analyze changes in the levels of plasma proteins of rats fed on a PUFAn-6 plus coenzyme Q(10)-based diet. This approach could give novel insights into the mechanisms of life span extension by dietary coenzyme Q(10) in the rat. Serum albumin, which decreases with aging in the rat, was significantly increased by coenzyme Q(10) supplementation both at 6 and 24 months. After depletion of the most abundant proteins by affinity chromatography, levels of less abundant plasma proteins were also studied by using 2D-electrophoresis and MALDI-TOF mass fingerprinting analysis. Our results have shown that lifelong dietary supplementation with coenzyme Q(10) induced significant decreases of plasma hemopexin, apolipoprotein H and inter-alpha-inhibitor H4P heavy chain (at both 6 and 24 months), preprohaptoglobin, fibrinogen gamma-chain precursor, and fetuin-like protein (at 6 months), and alpha-1-antitrypsin precursor and type II peroxiredoxin (at 24 months). On the other hand, coenzyme Q(10) supplementation resulted in significant increases of serine protease inhibitor 3, vitamin D-binding protein (at 6 months), and Apo A-I (at 24 months). Our results support a beneficial role of dietary coenzyme Q(10) decreasing oxidative stress and cardiovascular risk, and modulating inflammation during aging.


Assuntos
Suplementos Nutricionais , Longevidade/efeitos dos fármacos , Longevidade/fisiologia , Proteoma/efeitos dos fármacos , Proteoma/metabolismo , Ubiquinona/análogos & derivados , Animais , Proteínas Sanguíneas/efeitos dos fármacos , Proteínas Sanguíneas/metabolismo , Doenças Cardiovasculares/prevenção & controle , Coenzimas/administração & dosagem , Coenzimas/sangue , Eletroforese em Gel Bidimensional , Inflamação/prevenção & controle , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Ubiquinona/administração & dosagem , Ubiquinona/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...