Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Br J Nutr ; 131(8): 1352-1361, 2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38155410

RESUMEN

This study is aimed to evaluate the effect and underling mechanism of dietary supplementation with pyrroloquinoline quinone (PQQ) disodium on improving inflammatory liver injury in piglets challenged with lipopolysaccharide (LPS). A total of seventy-two crossbred barrows were allotted into four groups as follows: the CTRL group (basal diet + saline injection); the PQQ group (3 mg/kg PQQ diet + saline injection); the CTRL + LPS group (basal diet + LPS injection) and the PQQ + LPS group (3 mg/kg PQQ diet + LPS injection). On days 7, 11 and 14, piglets were challenged with LPS or saline. Blood was sampled at 4 h after the last LPS injection (day 14), and then the piglets were slaughtered and liver tissue was harvested. The results showed that the hepatic morphology was improved in the PQQ + LPS group compared with the CTRL + LPS group. PQQ supplementation decreased the level of serum inflammatory factors, aspartate aminotransferase and alanine transaminase, and increased the HDL-cholesterol concentration in piglets challenged with LPS; piglets in the PQQ + LPS group had lower liver mRNA level of inflammatory factors and protein level of α-smooth muscle actin than in the CTRL + LPS group. Besides, mRNA expression of STAT3/TGF-ß1 pathway and protein level of p-STAT3(Tyr 705) were decreased, and mRNA level of PPARα and protein expression of p-AMPK in liver were increased in the PQQ + LPS group compared with the CTRL + LPS group (P < 0·05). In conclusion, dietary supplementation with PQQ alleviated inflammatory liver injury might partly via inhibition of the STAT3/TGF-ß1 pathway in piglets challenged with LPS.


Asunto(s)
Suplementos Dietéticos , Lipopolisacáridos , Animales , Porcinos , Cofactor PQQ/farmacología , Cofactor PQQ/uso terapéutico , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Hígado/metabolismo , ARN Mensajero/metabolismo
2.
Aging Cell ; 22(9): e13912, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37365714

RESUMEN

Age-related osteoporosis is associated with increased oxidative stress and cellular senescence. Pyrroloquinoline quinone (PQQ) is a water-soluble vitamin-like compound that has strong antioxidant capacity; however, the effect and underlying mechanism of PQQ on aging-related osteoporosis remain unclear. The purpose of this study was to investigate whether dietary PQQ supplementation can prevent osteoporosis caused by natural aging, and the potential mechanism underlying PQQ antioxidant activity. Here, we found that when 6-month-old or 12-month-old wild-type mice were supplemented with PQQ for 12 months or 6 months, respectively, PQQ could prevent age-related osteoporosis in mice by inhibiting osteoclastic bone resorption and stimulating osteoblastic bone formation. Mechanistically, pharmmapper screening and molecular docking studies revealed that PQQ appears to bind to MCM3 and reduces its ubiquitination-mediated degradation; stabilized MCM3 then competes with Nrf2 for binding to Keap1, thus activating Nrf2-antioxidant response element (ARE) signaling. PQQ-induced Nrf2 activation inhibited bone resorption through increasing stress response capacity and transcriptionally upregulating fibrillin-1 (Fbn1), thus reducing Rankl production in osteoblast-lineage cells and decreasing osteoclast activation; as well, bone formation was stimulated by inhibiting osteoblastic DNA damage and osteocyte senescence. Furthermore, Nrf2 knockout significantly blunted the inhibitory effects of PQQ on oxidative stress, on increased osteoclast activity and on the development of aging-related osteoporosis. This study reveals the underlying mechanism of PQQ's strong antioxidant capacity and provides evidence for PQQ as a potential agent for clinical prevention and treatment of natural aging-induced osteoporosis.


Asunto(s)
Resorción Ósea , Osteoporosis , Ratones , Animales , Antioxidantes/metabolismo , Cofactor PQQ/farmacología , Cofactor PQQ/metabolismo , Cofactor PQQ/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Regulación hacia Arriba , Fibrilina-1/metabolismo , Simulación del Acoplamiento Molecular , Estrés Oxidativo , Envejecimiento , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Resorción Ósea/tratamiento farmacológico
3.
High Alt Med Biol ; 23(3): 249-263, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35384739

RESUMEN

Kushwaha, Asha D., and Deepika Saraswat. A nanocurcumin and pyrroloquinoline quinone formulation prevents hypobaric hypoxia-induced skeletal muscle atrophy by modulating NF-κB signaling pathway. High Alt Med Biol. 23:249-263, 2022. Background: Hypobaric hypoxia (HH)-induced deleterious skeletal muscle damage depends on exposure time and availability of oxygen at cellular level, which eventually can limit human work performance at high altitude (HA). Despite the advancements made in pharmacological (performance enhancer, antioxidants) and nonpharmacological therapeutics (acclimatization strategies), only partial success has been achieved in improving physical performance at HA. A distinctive combination of nanocurcumin (NC) and pyrroloquinoline quinone (PQQ) has been formulated (named NCF [nanocurcumin formulation], Indian patent No. 302877) in our laboratory, and has proven very promising in improving cardiomyocyte adaptation to chronic HH. We hypothesized that NCF might improve skeletal muscle adaptation and could be a performance enhancer at HA. Material and Methods: Adult Sprague-Dawley rats (220 ± 10 g) were divided into five groups (n = 6/group): normoxia vehicle control, hypoxia vehicle control, hypoxia NCF, hypoxia NC, and hypoxia PQQ. All the animals (except those in normoxia) were exposed to simulated HH in a chamber at temperature 22°C ± 2°C, humidity 50% ± 5%, altitude 25,000 ft for 1, 3, or 7 days. After completion of the stipulated exposure time, gastrocnemius and soleus muscles were excised from animals for further analysis. Results: Greater lengths of hypoxic exposure caused progressively increased muscle ring finger-1 (MuRF-1; p < 0.01) expression and calpain activation (0.56 ± 0.05 vs. 0.13 ± 0.02 and 0.44 ± 0.03 vs. 0.12 ± 0.021) by day 7, respectively in the gastrocnemius and soleus muscles. Myosin heavy chain type I (slow oxidative) fibers significantly (p > 0.01) decreased in gastrocnemius (>50%) and soleus (>46%) muscles by the seventh day of exposure. NCF supplementation showed (p ≤ 0.05) tremendous improvement in skeletal muscle acclimatization through effective alleviation of oxidative damage, and changes in calpain activity and atrophic markers at HA compared with hypoxia control or treatment alone with NC/PQQ. Conclusion: Thus, NCF-mediated anti-oxidative, anti-inflammatory effects lead to decreased proteolysis resulting in mitigated skeletal muscle atrophy under HH.


Asunto(s)
FN-kappa B , Cofactor PQQ , Animales , Atrofia/metabolismo , Calpaína/metabolismo , Calpaína/uso terapéutico , Humanos , Hipoxia/tratamiento farmacológico , Músculo Esquelético/metabolismo , FN-kappa B/metabolismo , FN-kappa B/uso terapéutico , Cofactor PQQ/metabolismo , Cofactor PQQ/uso terapéutico , Ratas , Ratas Sprague-Dawley , Transducción de Señal
4.
Reprod Sci ; 29(2): 578-585, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34542890

RESUMEN

Preeclampsia (PE) is a pregnancy complication commonly characterized by high blood pressure. Although it is generally believed that the placenta is the root cause of PE, the exact pathogenesis is unknown; consequently, there is no standard clinical treatment. Therefore, it is necessary to explore new therapeutic drugs. Several studies have reported that pyrroloquinoline quinone (PQQ) exhibits anti-inflammatory and antioxidative effects. The purpose of this study was to investigate the protective effect of PQQ diet supplementation on PE-like rat models. L-NAME induced PE-like model rats were intraperitonially administrated with PQQ. The results showed that PQQ significantly improved clinical manifestations and pregnancy outcomes of PE-like rats. The levels of related inflammatory and antioxidant markers were also significantly reversed. A mechanism study showed that PQQ may achieve the above therapeutic effects by inhibiting NF-κB and promoting Nrf2 antioxidant pathways. In conclusion, we showed the protective effect of PQQ on PE-like model rats, by improving anti-inflammation and antioxidation effect through the NF-κB-Nrf2 pathway.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , NG-Nitroarginina Metil Éster/farmacología , Cofactor PQQ/uso terapéutico , Preeclampsia/tratamiento farmacológico , Animales , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Inyecciones Intraperitoneales , Estrés Oxidativo/efectos de los fármacos , Preeclampsia/inducido químicamente , Embarazo , Ratas , Ratas Sprague-Dawley
5.
Int J Biol Sci ; 15(1): 58-68, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30662347

RESUMEN

Accumulating studies have shown that oxidative stress increases with aging, which is related to the pathophysiology of postmenopausal osteoporosis. Pyrroloquinoline quinone (PQQ) is a natural anti-oxidant with anti-oxidative and anti-aging effects. However, it is unclear whether PQQ has a protective role against estrogen deficiency-induced osteoporosis. Here, we evaluated the efficacy of PQQ on bone mineral density, bone microarchitecture, bone turnover and biomechanical strength in ovariectomy (OVX)-induced osteoporosis mouse model. Although dietary PQQ supplement did not affect serum E2 levels and uterine weight in OVX mice, it could prevent OVX-induced bone loss and improve bone strength by inhibiting oxidative stress, osteocyte senescence and senescence-associated secretory phenotype (SASP), subsequently promoting osteoblastic bone formation and inhibiting osteoclastic bone resorption, which was comparable to treatment with exogenous estrogen. The results from our study provide experimental evidence for the clinical use of PQQ to prevent estrogen deficiency-induced osteoporosis.


Asunto(s)
Estrógenos/deficiencia , Osteoporosis/etiología , Osteoporosis/prevención & control , Cofactor PQQ/uso terapéutico , Animales , Senescencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Ratones , Ratones Endogámicos C57BL , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Osteoporosis/metabolismo , Estrés Oxidativo/efectos de los fármacos
6.
J Cereb Blood Flow Metab ; 37(1): 69-84, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26661224

RESUMEN

Synaptic bioenergetic deficiencies may be associated with early Alzheimer's disease (AD). To explore this concept, we assessed pre-synaptic mitochondrial function in hemizygous (+/-)TgMcGill-R-Thy1-APP rats. The low burden of Aß and the wide array of behavioral and cognitive impairments described in 6-month-old hemizygous TgMcGill-R-Thy1-APP rats (Tg(+/-)) support their use to investigate synaptic bioenergetics deficiencies described in subjects with early Alzheimer's disease (AD). In this report, we show that pre-synaptic mitochondria from Tg(+/-) rats evidence a decreased respiratory control ratio and spare respiratory capacity associated with deficits in complex I enzymatic activity. Cognitive impairments were prevented and bioenergetic deficits partially reversed when Tg(+/-) rats were fed a nutritionally complete diet from weaning to 6-month-old supplemented with pyrroloquinoline quinone, a mitochondrial biogenesis stimulator with antioxidant and neuroprotective effects. These results provide evidence that, as described in AD brain and not proven in Tg mice models with AD-like phenotype, the mitochondrial bioenergetic capacity of synaptosomes is not conserved in the Tg(+/-) rats. This animal model may be suitable for understanding the basic biochemical mechanisms involved in early AD.


Asunto(s)
Enfermedad de Alzheimer/etiología , Disfunción Cognitiva/etiología , Metabolismo Energético , Sinaptosomas/metabolismo , Enfermedad de Alzheimer/dietoterapia , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Disfunción Cognitiva/metabolismo , Dietoterapia , Modelos Animales de Enfermedad , Complejo I de Transporte de Electrón/metabolismo , Mitocondrias/metabolismo , Cofactor PQQ/uso terapéutico , Ratas , Ratas Transgénicas
7.
FASEB J ; 31(4): 1434-1448, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28007783

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is widespread in adults and children. Early exposure to maternal obesity or Western-style diet (WD) increases steatosis and oxidative stress in fetal liver and is associated with lifetime disease risk in the offspring. Pyrroloquinoline quinone (PQQ) is a natural antioxidant found in soil, enriched in human breast milk, and essential for development in mammals. We investigated whether a supplemental dose of PQQ, provided prenatally in a mouse model of diet-induced obesity during pregnancy, could protect obese offspring from progression of NAFLD. PQQ treatment given pre- and postnatally in WD-fed offspring had no effect on weight gain but increased metabolic flexibility while reducing body fat and liver lipids, compared with untreated obese offspring. Indices of NAFLD, including hepatic ceramide levels, oxidative stress, and expression of proinflammatory genes (Nos2, Nlrp3, Il6, and Ptgs2), were decreased in WD PQQ-fed mice, concomitant with increased expression of fatty acid oxidation genes and decreased Pparg expression. Notably, these changes persisted even after PQQ withdrawal at weaning. Our results suggest that supplementation with PQQ, particularly during pregnancy and lactation, protects offspring from WD-induced developmental programming of hepatic lipotoxicity and may help slow the advancing epidemic of NAFLD in the next generation.-Jonscher, K. R., Stewart, M. S., Alfonso-Garcia, A., DeFelice, B. C., Wang, X. X., Luo, Y., Levi, M., Heerwagen, M. J. R., Janssen, R. C., de la Houssaye, B. A., Wiitala, E., Florey, G., Jonscher, R. L., Potma, E. O., Fiehn, O. Friedman, J. E. Early PQQ supplementation has persistent long-term protective effects on developmental programming of hepatic lipotoxicity and inflammation in obese mice.


Asunto(s)
Antioxidantes/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Obesidad/complicaciones , Cofactor PQQ/uso terapéutico , Efectos Tardíos de la Exposición Prenatal/prevención & control , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Ceramidas/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Femenino , Interleucina-6/genética , Interleucina-6/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/etiología , Obesidad/tratamiento farmacológico , Obesidad/etiología , Estrés Oxidativo , PPAR gamma/metabolismo , Cofactor PQQ/administración & dosificación , Cofactor PQQ/farmacología , Embarazo , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/etiología
8.
Biochem Biophys Res Commun ; 457(4): 507-13, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25580010

RESUMEN

Regeneration and functional reconstruction of peripheral nerve defects remained a significant clinical challenge. Nerve guide conduits, with seed cells or neurotrophic factors (NTFs), had been widely used to improve the repair and regeneration of injured peripheral nerve. Pyrroloquinoline quinone (PQQ) was an antioxidant that can stimulate nerve growth factors (NGFs) synthesis and accelerate the Schwann cells (SCs) proliferation and growth. In present study, three kinds of nerve guide conduits were constructed: one from cellulose/SPI hollow tube (CSC), another from CSC combined with SCs (CSSC), and the third one from CSSC combined with PQQ (CSSPC), respectively. And then they were applied to bridge and repair the sciatic nerve defect in rats, using autograft as control. Effects of different nerve guide conduits on the nerve regeneration were comparatively evaluated by general analysis, sciatic function index (SFI) and histological analysis (HE and TEM). Newly-formed regenerative nerve fibers were observed and running through the transparent nerve guide conduits 12 weeks after surgery. SFI results indicated that the reconstruction of motor function in CSSPC group was better than that in CSSC and CSC groups. HE images from the cross-sections and longitudinal-sections of the harvested regenerative nerve indicated that regenerative nerve fibers had been formed and accompanied with new blood vessels and matrix materials in the conduits. TEM images also showed that lots of fresh myelinated and non-myelinated nerve fibers had been formed. Parts of vacuolar, swollen and abnormal axons occurred in CSC and CSSC groups, while the vacuolization and swell of axons was the least serious in CSSPC group. These results indicated that CSSPC group had the most ability to repair and reconstruct the nerve structure and functions due to the comprehensive contributions from hollow CSC tube, SCs and PQQ. As a result, the CSSPC may have the potential for the applications as nerve guide conduits in the field of nerve tissue engineering.


Asunto(s)
Antioxidantes/uso terapéutico , Celulosa/química , Regeneración Nerviosa/efectos de los fármacos , Cofactor PQQ/uso terapéutico , Células de Schwann/citología , Nervio Ciático/fisiología , Proteínas de Soja/química , Andamios del Tejido/química , Animales , Antioxidantes/administración & dosificación , Regeneración Tisular Dirigida , Masculino , Cofactor PQQ/administración & dosificación , Ratas , Ratas Sprague-Dawley , Células de Schwann/efectos de los fármacos , Nervio Ciático/lesiones , Nervio Ciático/patología , Nervio Ciático/ultraestructura , Ingeniería de Tejidos
9.
PLoS One ; 6(7): e21779, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21814553

RESUMEN

We have reported that pyrroloquinoline quinone (PQQ) improves reproduction, neonatal development, and mitochondrial function in animals by mechanisms that involve mitochondrial related cell signaling pathways. To extend these observations, the influence of PQQ on energy and lipid relationships and apparent protection against ischemia reperfusion injury are described herein. Sprague-Dawley rats were fed a nutritionally complete diet with PQQ added at either 0 (PQQ-) or 2 mg PQQ/Kg diet (PQQ+). Measurements included: 1) serum glucose and insulin, 2) total energy expenditure per metabolic body size (Wt(3/4)), 3) respiratory quotients (in the fed and fasted states), 4) changes in plasma lipids, 5) the relative mitochondrial amount in liver and heart, and 6) indices related to cardiac ischemia. For the latter, rats (PQQ- or PQQ+) were subjected to left anterior descending occlusions followed by 2 h of reperfusion to determine PQQ's influence on infarct size and myocardial tissue levels of malondialdehyde, an indicator of lipid peroxidation. Although no striking differences in serum glucose, insulin, and free fatty acid levels were observed, energy expenditure was lower in PQQ- vs. PQQ+ rats and energy expenditure (fed state) was correlated with the hepatic mitochondrial content. Elevations in plasma di- and triacylglyceride and ß-hydroxybutryic acid concentrations were also observed in PQQ- rats vs. PQQ+ rats. Moreover, PQQ administration (i.p. at 4.5 mg/kg BW for 3 days) resulted in a greater than 2-fold decrease in plasma triglycerides during a 6-hour fast than saline administration in a rat model of type 2 diabetes. Cardiac injury resulting from ischemia/reperfusion was more pronounced in PQQ- rats than in PQQ+ rats. Collectively, these data demonstrate that PQQ deficiency impacts a number of parameters related to normal mitochondrial function.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Lípidos/análisis , Mitocondrias/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Cofactor PQQ/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/prevención & control , Modelos Animales de Enfermedad , Glucosa/metabolismo , Corazón/efectos de los fármacos , Pruebas de Función Cardíaca , Peroxidación de Lípido/efectos de los fármacos , Malondialdehído/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Estado Nutricional , Ratas , Ratas Sprague-Dawley , Tasa de Supervivencia
10.
J Nutr Sci Vitaminol (Tokyo) ; 55(5): 389-93, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19926923

RESUMEN

In order to verify whether vitamin E improves the cognitive impairment induced through aging, aged rats fed a vitamin E-supplemented diet had their learning and memory functions assessed in comparison with the aged rats fed a normal diet using a Morris water maze test. Although normal aged rats showed very poor learning ability concerning the place of a platform in the water maze apparatus, the aged rats fed the vitamin E-supplemented diet learned the place with a marked speed in only 5 trials. After old animals showed the maximum learning ability, they were kept in a normal atmosphere for 48 h without a trial followed by an assessment of their memory function using the same apparatus. The vitamin E-supplementation to aged rats resulted in marked retention of their maximum memory function, although normal aged rats showed a significant memory loss of about 60%. Pyrroloquinoline quinone (PQQ), which increases in the production of nerve growth factor, and protects neurons, had a similar effect on cognitive function to that of vitamin E in the aged rats. These results suggest that vitamin E may improve cognitive deficit caused through aging by not only its neuro-protecting effect but an antioxidant efficacy.


Asunto(s)
Antioxidantes/uso terapéutico , Suplementos Dietéticos , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Vitamina E/uso terapéutico , Envejecimiento/psicología , Animales , Antioxidantes/farmacología , Cognición/efectos de los fármacos , Masculino , Fármacos Neuroprotectores/farmacología , Cofactor PQQ/farmacología , Cofactor PQQ/uso terapéutico , Ratas , Ratas Wistar , Vitamina E/farmacología
11.
Altern Med Rev ; 14(3): 268-77, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19803551

RESUMEN

Pyrroloquinoline quinone (PQQ) is a novel biofactor for which a proposition can be made for physiological importance. PQQ was first recognized as an enzyme cofactor in bacteria. It has recently been tentatively identified as a component of interstellar dust. Thus, PQQ may have been present throughout early biological conception and evolution. PQQ is also a potent plant growth factor. Consequently, for animals and humans, there has been constant exposure to PQQ. In animals, PQQ is reported to participate in a range of biological functions with apparent survival benefits (e.g., improved neonatal growth and reproductive performance). There are also benefits from PQQ supplementation related to cognitive, immune, and antioxidant functions, as well as protection from cardiac and neurological ischemic events. Although PQQ is not currently viewed as a vitamin, its involvement in cell signaling pathways, particularly those important to mitochondriogenesis in experimental animal models, may eventually provide a rationale for defining PQQ as vital to life. For humans, such evidence suggests there may be similar parallels or benefits from improving PQQ status.


Asunto(s)
Antioxidantes/farmacología , Factores Quimiotácticos/farmacología , Fármacos Neuroprotectores/farmacología , Cofactor PQQ/farmacología , Animales , Antioxidantes/uso terapéutico , Proliferación Celular/efectos de los fármacos , Factores Quimiotácticos/uso terapéutico , Cognición/efectos de los fármacos , Cardiopatías/prevención & control , Humanos , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Cofactor PQQ/uso terapéutico , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA