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








Base de dados
Intervalo de ano de publicação
1.
J Appl Toxicol ; 44(2): 235-244, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37650462

RESUMO

Gentamicin (GM) is one of the commonly used antibiotics in the aminoglycoside class but is ototoxic, which constantly impacts the quality of human life. Pyrroloquinoline quinone (PQQ) as a redox cofactor produced by bacteria was found in soil and foods that exert an antioxidant and redox modulator. It is well documented that the PQQ can alleviate inflammatory responses and cytotoxicity. However, our understanding of PQQ in ototoxicity remains unclear. We reported that PQQ could protect against GM-induced ototoxicity in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells in vitro. To evaluate reactive oxygen species (ROS) production and mitochondrial function, ROS and JC-1 staining, oxygen consumption rate (OCR), and extracellular acidification rate (ECAR) measurements in living cells, mitochondrial dynamics analysis was performed. GM-mediated damage was performed by reducing the production of ROS and inhibiting mitochondria biogenesis and dynamics. PQQ ameliorated the cellular oxidative stress and recovered mitochondrial membrane potential, facilitating the recovery of mitochondrial biogenesis and dynamics. Our in vitro findings improve our understanding of the GM-induced ototoxicity with therapeutic implications for PQQ.


Assuntos
Gentamicinas , Ototoxicidade , Humanos , Gentamicinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Cofator PQQ/farmacologia , Cofator PQQ/uso terapêutico , Cofator PQQ/metabolismo , Ototoxicidade/etiologia , Ototoxicidade/prevenção & controle , Ototoxicidade/metabolismo , Células Ciliadas Auditivas/metabolismo , Antibacterianos/metabolismo , Apoptose
2.
Br J Nutr ; 131(8): 1352-1361, 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38155410

RESUMO

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.


Assuntos
Suplementos Nutricionais , Lipopolissacarídeos , Animais , Suínos , Cofator PQQ/farmacologia , Cofator PQQ/uso terapêutico , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Fígado/metabolismo , RNA Mensageiro/metabolismo
3.
Fluids Barriers CNS ; 20(1): 84, 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37981683

RESUMO

BACKGROUND: Folates (Vitamin B9) are critical for normal neurodevelopment and function, with transport mediated by three major pathways: folate receptor alpha (FRα), proton-coupled folate transporter (PCFT), and reduced folate carrier (RFC). Cerebral folate uptake primarily occurs at the blood-cerebrospinal fluid barrier (BCSFB) through concerted actions of FRα and PCFT, with impaired folate transport resulting in the neurological disorder cerebral folate deficiency (CFD). Increasing evidence suggests that disorders associated with CFD also present with neuroinflammation, oxidative stress, and mitochondrial dysfunction, however the role of brain folate deficiency in inducing these abnormalities is not well-understood. Our laboratory has identified the upregulation of RFC by nuclear respiratory factor 1 (NRF-1) at the blood-brain barrier (BBB) once indirectly activated by the natural compound pyrroloquinoline quinone (PQQ). PQQ is also of interest due to its anti-inflammatory, antioxidant, and mitochondrial biogenesis effects. In this study, we examined the effects of folate deficiency and PQQ treatment on inflammatory and oxidative stress responses, and changes in mitochondrial function. METHODS: Primary cultures of mouse mixed glial cells exposed to folate-deficient (FD) conditions and treated with PQQ were analyzed for changes in gene expression of the folate transporters, inflammatory markers, oxidative stress markers, and mitochondrial DNA (mtDNA) content through qPCR analysis. Changes in cellular reactive oxygen species (ROS) levels were analyzed in vitro through a DCFDA assay. Wildtype (C57BL6/N) mice exposed to FD (0 mg/kg folate), or control (2 mg/kg folate) diets underwent a 10-day (20 mg/kg/day) PQQ treatment regimen and brain tissues were collected and analyzed. RESULTS: Folate deficiency resulted in increased expression of inflammatory and oxidative stress markers in vitro and in vivo, with increased cellular ROS levels observed in mixed glial cells as well as a reduction of mitochondrial DNA (mtDNA) content observed in FD mixed glial cells. PQQ treatment was able to reverse these changes, while increasing RFC expression through activation of the PGC-1α/NRF-1 signaling pathway. CONCLUSION: These results demonstrate the effects of brain folate deficiency, which may contribute to the neurological deficits commonly seen in disorders of CFD. PQQ may represent a novel treatment strategy for disorders associated with CFD, as it can increase folate uptake, while in parallel reversing many abnormalities that arise with brain folate deficiency.


Assuntos
Encéfalo , Cofator PQQ , Animais , Camundongos , Cofator PQQ/farmacologia , Cofator PQQ/uso terapêutico , Espécies Reativas de Oxigênio , Ácido Fólico/farmacologia , DNA Mitocondrial
4.
Aging Cell ; 22(9): e13912, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37365714

RESUMO

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.


Assuntos
Reabsorção Óssea , Osteoporose , Camundongos , Animais , Antioxidantes/metabolismo , Cofator PQQ/farmacologia , Cofator PQQ/metabolismo , Cofator PQQ/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Regulação para Cima , Fibrilina-1/metabolismo , Simulação de Acoplamento Molecular , Estresse Oxidativo , Envelhecimento , Osteoporose/tratamento farmacológico , Osteoporose/metabolismo , Reabsorção Óssea/tratamento farmacológico
5.
J Dev Orig Health Dis ; 14(3): 321-324, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36861270

RESUMO

BACKGROUND: Intrauterine growth restriction (IUGR) exerts a negative impact on developing cardiomyocytes and emerging evidence suggests activation of oxidative stress pathways plays a key role in this altered development. Here, we provided pregnant guinea pig sows with PQQ, an aromatic tricyclic o-quinone that functions as a redox cofactor antioxidant, during the last half of gestation as a potential antioxidant intervention for IUGR-associated cardiomyopathy. METHODS: Pregnant guinea pig sows were randomly assigned to receive PQQ or placebo at mid gestation and fetuses were identified as spontaneous IUGR (spIUGR) or normal growth (NG) near term yielding four cohorts: NG ± PQQ and spIUGR ± PQQ. Cross sections of fetal left and right ventricles were prepared and cardiomyocyte number, collagen deposition, proliferation (Ki67) and apoptosis (TUNEL) were analyzed. RESULTS: Cardiomyocyte endowment was reduced in spIUGR fetal hearts when compared to NG; however, PQQ exerted a positive effect on cardiomyocyte number in spIUGR hearts. Cardiomyocytes undergoing proliferation and apoptosis were more common in spIUGR ventricles when compared with NG animals, which was significantly reduced with PQQ supplementation. Similarly, collagen deposition was increased in spIUGR ventricles and was partially rescued in PQQ-treated spIUGR animals. CONCLUSION: The negative influence of spIUGR on cardiomyocyte number, apoptosis, and collagen deposition during parturition can be suppressed by antenatal administration of PQQ to pregnant sows. These data identify a novel therapeutic intervention for irreversible spIUGR-associated cardiomyopathy.


Assuntos
Retardo do Crescimento Fetal , Miócitos Cardíacos , Animais , Feminino , Cobaias , Gravidez , Antioxidantes , Retardo do Crescimento Fetal/tratamento farmacológico , Retardo do Crescimento Fetal/metabolismo , Oxirredução , Cofator PQQ/farmacologia , Cofator PQQ/uso terapêutico
6.
Mediators Inflamm ; 2022: 1267841, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36345503

RESUMO

The current asthma therapies are inadequate for many patients with severe asthma. Pyrroloquinoline quinone (PQQ) is a naturally-occurring redox cofactor and nutrient that can exert a multitude of physiological effects, including anti-inflammatory and antioxidative effects. We sought to explore the effects of PQQ on allergic airway inflammation and reveal the underlying mechanisms. In vitro, the effects of PQQ on the secretion of epithelial-derived cytokines by house dust mite- (HDM-) incubated 16-HBE cells and on the differentiation potential of CD4+ T cells were investigated. In vivo, PQQ was administered to mice with ovalbumin- (OVA-) induced asthma, and lung pathology and inflammatory cell infiltration were assessed. The changes in T cell subsets and signal transducers and activators of transcription (STATs) were evaluated by flow cytometry. Pretreatment with PQQ significantly decreased HDM-stimulated thymic stromal lymphopoietin (TSLP) production in a dose-dependent manner in 16-HBE cells and inhibited Th2 cell differentiation in vitro. Treatment with PQQ significantly reduced bronchoalveolar lavage fluid (BALF) inflammatory cell counts in the OVA-induced mouse model. PQQ administration also changed the secretion of IFN-γ and IL-4 as well as the percentages of Th1, Th2, Th17, and Treg cells in the peripheral blood and lung tissues, along with inhibition the phosphorylation of STAT1, STAT3, and STAT6 while promoting that of STAT4 in allergic airway inflammation model mice. PQQ can alleviate allergic airway inflammation in mice by improving the immune microenvironment and regulating the JAK-STAT signaling pathway. Our findings suggest that PQQ has great potential as a novel therapeutic agent for inflammatory diseases, including asthma.


Assuntos
Asma , Cofator PQQ , Animais , Camundongos , Asma/metabolismo , Líquido da Lavagem Broncoalveolar , Citocinas/metabolismo , Modelos Animais de Doenças , Inflamação/metabolismo , Pulmão/metabolismo , Camundongos Endogâmicos BALB C , Ovalbumina , Cofator PQQ/farmacologia , Cofator PQQ/uso terapêutico , Cofator PQQ/metabolismo , Transdução de Sinais , Células Th2 , Fatores de Transcrição STAT/metabolismo , Janus Quinases
7.
Pulm Pharmacol Ther ; 76: 102156, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36030026

RESUMO

Excessive proliferation of pulmonary artery smooth muscle cells (PASMCs) and endothelial cells (PAECs), inflammation, as well as mitochondrial and metabolic dysregulation, contributes to the development of pulmonary hypertension (PH). Pyrroloquinoline quinone (PQQ), a potent natural antioxidant with anti-diabetic, neuroprotective, and cardioprotective properties, is known to promote mitochondrial biogenesis. However, its effect on cellular proliferation, apoptosis resistance, mitochondrial and metabolic alterations associated with PH remains unexplored. The current study was designed to investigate the effect of PQQ in the treatment of PH. Human pulmonary artery smooth muscle cells (HPASMCs), endothelial cells (PAECs), and primary cultured cardiomyocytes were subjected to hypoxia to induce PH-like phenotype. Furthermore, Sprague Dawley (SD) rats injected with monocrotaline (MCT) (60 mg/kg, SC, once) progressively developed pulmonary hypertension. PQQ treatment (2 mg/kg, PO, for 35 days) attenuated cellular proliferation and promoted apoptosis via a mitochondrial-dependent pathway. Furthermore, PQQ treatment in HPASMCs prevented mitochondrial and metabolic dysfunctions, improved mitochondrial bioenergetics while preserving respiratory complexes, and reduced insulin resistance. In addition, PQQ treatment (preventive and curative) significantly attenuated the increase in right ventricle pressure and hypertrophy as well as reduced endothelial dysfunction and pulmonary artery remodeling in MCT-treated rats. PQQ also prevented cardiac fibrosis and improved cardiac functions as well as reduced inflammation in MCT-treated rats. Altogether, the above findings demonstrate that PQQ can attenuate mitochondrial as well as metabolic abnormalities in PASMCs and also prevent the development of PH in MCT treated rats; hence PQQ may act as a potential therapeutic agent for the treatment of PH.


Assuntos
Hipertensão Pulmonar , Animais , Células Endoteliais , Humanos , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/tratamento farmacológico , Inflamação/tratamento farmacológico , Monocrotalina , Cofator PQQ/metabolismo , Cofator PQQ/farmacologia , Cofator PQQ/uso terapêutico , Artéria Pulmonar , Ratos , Ratos Sprague-Dawley
8.
Biomed Pharmacother ; 150: 112998, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35489281

RESUMO

Diabetic nephropathy (DN), which is characterized by renal fibrosis, is a major complication of diabetes, a disease that afflicted more than 460 million people worldwide in 2019. Pyroptosis is an essential signaling pathway in DN-related injuries, such as renal fibrosis. Pyrroloquinoline quinone (PQQ) is a naturally occurring bioactive compound that protects human kidney 2 (HK-2) cells from oxidative stress-induced damage caused by high glucose concentrations. However, the nature and underlying mechanism of the effect of PQQ on DN-related renal fibrosis remains unclear. In this study, we evaluated whether PQQ has potential protective effects against renal fibrosis due to DN by establishing type 1 diabetes in mice via streptozotocin treatment and then inhibiting their pyroptosis signaling pathway. We found that compared to control mice, the area of renal fibrosis and injury were significantly increased in diabetic mice, and this was accompanied by increased levels of expression of collagen Ⅰ and transforming growth factor-ß1; increased concentrations of the inflammatory cytokines, interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α; and activation of the pyroptosis pathway components nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), caspase-1, IL-1ß, and IL-18. All of these changes were reversed by PQQ treatment. Analogously, we treated cultured HK-2 cells with a high concentration of glucose (35 mmol/L), which caused these cells to exhibit significantly increased concentrations of reactive oxygen species (ROS), phosphorylated (p)-nuclear factor kappa B (NF-κB), p-IkappaB, NLRP3, caspase-1, IL-1ß, and IL-18, and the loss of mitochondrial transmembrane potential. However, PQQ treatment significantly blunted these effects. In conclusion, in this study we demonstrated that PQQ attenuates renal fibrosis by alleviating mitochondrial dysfunction, reducing ROS production, and inhibiting the activation of the NF-κB/pyroptosis pathway under conditions of DN and hyperglycemia.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Animais , Caspase 1 , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Fibrose , Glucose/farmacologia , Humanos , Interleucina-18 , Rim , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Cofator PQQ/farmacologia , Cofator PQQ/uso terapêutico , Piroptose , Espécies Reativas de Oxigênio/metabolismo
9.
High Alt Med Biol ; 23(3): 249-263, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35384739

RESUMO

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.


Assuntos
NF-kappa B , Cofator PQQ , Animais , Atrofia/metabolismo , Calpaína/metabolismo , Calpaína/uso terapêutico , Humanos , Hipóxia/tratamento farmacológico , Músculo Esquelético/metabolismo , NF-kappa B/metabolismo , NF-kappa B/uso terapêutico , Cofator PQQ/metabolismo , Cofator PQQ/uso terapêutico , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
10.
Eur J Nutr ; 61(4): 1823-1836, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34997266

RESUMO

PURPOSE: Diabetic cardiomyopathy (DCM), a common complication of diabetes mellitus and is characterized by myocardial hypertrophy and myocardial fibrosis. Pyrroloquinoline quinone (PQQ), a natural nutrient, exerts strong protection against various myocardial diseases. Pyroptosis, a type of inflammation-related programmed cell death, is vital to the development of DCM. However, the protective effects of PQQ against DCM and the associated mechanisms are not clear. This study aimed to investigate whether PQQ protected against DCM and to determine the underlying molecular mechanism. METHODS: Diabetes was induced in mice by intraperitoneal injection of streptozotocin, after which the mice were administered PQQ orally (10, 20, or 40 mg/kg body weight/day) for 12 weeks. AC16 human myocardial cells were divided into the following groups and treated accordingly: control (5.5 mmol/L glucose), high glucose (35 mmol/L glucose), and HG + PQQ groups (1 and 10 nmol/L PQQ). Cells were treated for 24 h. RESULTS: PQQ reduced myocardial hypertrophy and the area of myocardial fibrosis, which was accompanied by an increase in antioxidant function and a decrease in inflammatory cytokine levels. Moreover, myocardial hypertrophy-(ANP and BNP), myocardial fibrosis-(collagen I and TGF-ß1), and pyroptosis-related protein levels decreased in the PQQ treatment groups. Furthermore, PQQ abolished mitochondrial dysfunction and the activation of NF-κB/IκB, and decreased NLRP3 inflammation-mediated pyroptosis in AC16 cells under high-glucose conditions. CONCLUSION: PQQ improved DCM in diabetic mice by inhibiting NF-κB/NLRP3 inflammasome-mediated cell pyroptosis. Long-term dietary supplementation with PQQ may be greatly beneficial for the treatment of DCM. Diagram of the underlying mechanism of the effects of PQQ on DCM. PQQ inhibits ROS generation and NF-κB activation, which stimulates activation of the NLRP3 inflammasome and regulates the expression of caspase-1, IL-1ß, and IL-18. The up-regulated inflammatory cytokines trigger myocardial hypertrophy and cardiac fibrosis and promote the pathological process of DCM.


Assuntos
Diabetes Mellitus Experimental , Cardiomiopatias Diabéticas , Animais , Cardiomegalia , Diabetes Mellitus Experimental/complicações , Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/etiologia , Cardiomiopatias Diabéticas/metabolismo , Fibrose , Glucose , Inflamassomos/metabolismo , Inflamação/complicações , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Cofator PQQ/metabolismo , Cofator PQQ/farmacologia , Cofator PQQ/uso terapêutico , Piroptose , Transdução de Sinais
11.
Reprod Sci ; 29(2): 578-585, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34542890

RESUMO

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.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , NG-Nitroarginina Metil Éster/farmacologia , Cofator PQQ/uso terapêutico , Pré-Eclâmpsia/tratamento farmacológico , Animais , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Injeções Intraperitoneais , Estresse Oxidativo/efeitos dos fármacos , Pré-Eclâmpsia/induzido quimicamente , Gravidez , Ratos , Ratos Sprague-Dawley
12.
Biomed Pharmacother ; 139: 111598, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33895522

RESUMO

It is reported that oxidative stress plays a detrimental role in the process of bone fracture healing. And pyrroloquinoline quinone (PQQ) is used as antioxidant. However, there is no report about whether PQQ supplementation can promote fracture healing by eliminating oxidative stress. To investigate the protective effect of PQQ on fracture healing, open mid-diaphyseal femur fractures model were created in sham, ovariectomized (OVX) mice and PQQ-treated OVX mice. Our results confirmed that PQQ played a preventive and protective role in OVX-induced delay of bone fracture healing by inhibiting oxidative stress, subsequently promoting osteoblastic bone formation and inhibiting osteoclastic bone resorption. The findings of this study not only revealed the mechanism of PQQ supplementation in promoting fracture healing, but also provide experimental and theoretical basis for the clinical application of PQQ in the treatment of bone fracture.


Assuntos
Antioxidantes/uso terapêutico , Fêmur/lesões , Fraturas Ósseas/tratamento farmacológico , Cofator PQQ/uso terapêutico , Animais , Antioxidantes/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Estrogênios/deficiência , Feminino , Fêmur/diagnóstico por imagem , Consolidação da Fratura/efeitos dos fármacos , Fraturas Ósseas/diagnóstico por imagem , Camundongos Endogâmicos C57BL , Ovariectomia , Estresse Oxidativo/efeitos dos fármacos , Cofator PQQ/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Microtomografia por Raio-X
13.
Acta Pharmacol Sin ; 42(5): 665-678, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32860006

RESUMO

Mitochondrial dysfunction is considered to be one of the important pathogenesis in Parkinson's disease (PD). We previously showed that pyrroloquinoline quinone (PQQ) could protect SH-SY5Y cells and dopaminergic neurons from cytotoxicity and prevent mitochondrial dysfunction in rotenone-induced PD models. In the present study we investigated the mechanisms underlying the protective effects of PQQ in a mouse PD model, which was established by intraperitoneal injection of rotenone (3 mg·kg-1·d-1, ip) for 3 weeks. Meanwhile the mice were treated with PQQ (0.8, 4, 20 mg·kg-1·d-1, ip) right after rotenone injection for 3 weeks. We showed that PQQ treatment dose-dependently alleviated the locomotor deficits and nigral dopaminergic neuron loss in PD mice. Furthermore, PQQ treatment significantly diminished the reduction of mitochondria number and their pathological change in the midbrain. PQQ dose-dependently blocked rotenone-caused reduction in the expression of PGC-1α and TFAM, two key activators of mitochondrial gene transcription, in the midbrain. In rotenone-injured human neuroblastoma SH-SY5Y cells, PTMScan Direct analysis revealed that treatment with PQQ (100 µM) differentially regulated protein phosphorylation; the differentially expressed phosphorylated proteins included the signaling pathways related with adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) pathway. We conducted Western blot analysis and confirmed that AMPK was activated by PQQ both in PD mice and in rotenone-injured SH-SY5Y cells. Pretreatment with AMPK inhibitor dorsomorphin (4 µM) significantly attenuated the protective effect and mitochondrial biogenesis by PQQ treatment in rotenone-injured SH-SY5Y cells. Taken together, PQQ promotes mitochondrial biogenesis in rotenone-injured mice and SH-SY5Y cells via activation of AMPK signaling pathway.


Assuntos
Mitocôndrias/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Biogênese de Organelas , Cofator PQQ/uso terapêutico , Doença de Parkinson Secundária/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Locomoção/efeitos dos fármacos , Masculino , Camundongos Endogâmicos ICR , Proteínas do Tecido Nervoso/efeitos dos fármacos , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/enzimologia , Fosforilação/efeitos dos fármacos , Rotenona
14.
Molecules ; 25(19)2020 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-32977419

RESUMO

Neuroinflammation is a feature common to neurodegenerative diseases, such as Parkinson's disease (PD), which might be responsive to therapeutic intervention. Rotenone has been widely used to establish PD models by inducing mitochondrial dysfunction and inflammation. Our previous studies have reported that pyrroloquinoline quinone (PQQ), a naturally occurring redox cofactor, could prevent mitochondrial dysfunction in rotenone induced PD models by regulating mitochondrial functions. In the present study, we aimed to investigate the effect of PQQ on neuroinflammation and the mechanism involved. BV2 microglia cells were pre-treated with PQQ followed by rotenone incubation. The data showed that PQQ did not affect the cell viability of BV2 cells treated with rotenone, while the conditioned medium (CM) of BV2 cells pre-treated with PQQ significantly increased cell viability of SH-SY5Y cells. In rotenone-treated BV2 cells, PQQ dose-dependently decreased lactate dehydrogenase (LDH) release and suppressed the up-regulation of pro-inflammation factors, such as interleukin-1ß (IL-1ß), IL-6 and tumor necrosis factor-α (TNF-α) in the cultured media, as well as nitric oxide (NO) release induced by rotenone. PQQ pretreatment also increased the ratio of LC3-II/LC3-I and expression of Atg5 in BV2 cells stimulated with rotenone. Additionally, the autophagosome observed by transmission electron microscopy (TEM) and co-localization of mitochondria with lysosomes indicated that mitophagy was induced by PQQ in rotenone-injured BV2 cells, and the PINK1/parkin mediated mitophagy pathway was regulated by PQQ. Further, autophagy inhibitor, 3-methyladenine (3-MA), partially abolished the neuroprotective effect of PQQ and attenuated the inhibition of inflammation with PQQ pretreatment. Taken together, our data extend our understanding of the neuroprotective effect of PQQ against rotenone-induced injury and provide evidence that autophagy enhancement might be a novel therapeutic strategy for PD treatment.


Assuntos
Anti-Inflamatórios/farmacologia , Autofagia/efeitos dos fármacos , Microglia/citologia , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Cofator PQQ/farmacologia , Rotenona/efeitos adversos , Anti-Inflamatórios/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/patologia , Fármacos Neuroprotetores/uso terapêutico , Cofator PQQ/uso terapêutico
15.
Life Sci ; 256: 117901, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32504759

RESUMO

AIMS: Cyclophosphamide (CTX) is an effective anti-tumor and immunosuppressive agent, but it induces nephrotoxicity in clinical applications. The present study aimed to evaluate the protective effect of pyrroloquinoline quinone (PQQ) on CTX-induced nephrotoxicity. MAIN METHODS: We injected male ICR mice with CTX (80 mg/kg/day), and determined nephrotoxicity indices, MDA and antioxidant defenses, inflammatory cytokines, and the levels of main proteins in the Nrf2-HO-1 and NLRP3 signaling pathways. KEY FINDINGS: PQQ has significantly decreased the serum levels of creatinine and urea compared to Model group. When treated with PQQ, MDA, IL-1ß, IL-6, and TNF-α levels have decreased, and SOD, GSH-Px, and CAT activity have increased in the kidney tissues of CTX-induced mice. PQQ activated the Nrf2-mediated signaling pathway, as indicated by the increased expression of Nrf2, HO-1, GCLM, and NQO1. Moreover, PQQ inhibited the NLRP3 inflammatory pathway, as indicated by the reduced expression of NLRP3, ASC, and Caspase-1. SIGNIFICANCE: Our results suggest that PQQ protects against CTX-induced nephrotoxicity, probably by activating the Nrf2-mediated antioxidant pathway and inhibiting the NLRP3 inflammatory pathway.


Assuntos
Ciclofosfamida/efeitos adversos , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Cofator PQQ/uso terapêutico , Transdução de Sinais , Animais , Antioxidantes/metabolismo , Nitrogênio da Ureia Sanguínea , Peso Corporal/efeitos dos fármacos , Creatinina/metabolismo , Citocinas/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Rim/efeitos dos fármacos , Rim/enzimologia , Rim/patologia , Masculino , Malondialdeído/metabolismo , Camundongos Endogâmicos ICR , Modelos Biológicos , Tamanho do Órgão/efeitos dos fármacos , Cofator PQQ/química , Cofator PQQ/farmacologia
16.
Curr Stem Cell Res Ther ; 15(1): 32-36, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-30526470

RESUMO

Pyrroloquinoline Quinone (PQQ) is the third coenzyme found after niacinamide and flavone nucleotides and is widely present in microorganisms, plants, animals, and humans. PQQ can stimulate the growth of organisms and is very important for the growth, development and reproduction of animals. Owing to the inherent properties of PQQ as an antioxidant and redox modulator in various systems. In recent years, the role of PQQ in the field of osteoporosis and neuro injury has become a research hotspot. This article mainly discusses the derivatives, distribution of PQQ, in vitro models of osteoporosis and neuro injury, and the research progress of its mechanism of action. It provides new ideas in the study of osteoporosis and neuro injury.


Assuntos
Antioxidantes/uso terapêutico , Bactérias Gram-Positivas/fisiologia , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Sistema Nervoso/fisiopatologia , Osteoporose/tratamento farmacológico , Cofator PQQ/uso terapêutico , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Cofator PQQ/análogos & derivados
17.
Hum Exp Toxicol ; 39(2): 194-211, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31661991

RESUMO

Mono(2-ethylhexyl) phthalate (MEHP) is the main metabolite of di(2-ethylhexyl) phthalate (DEHP) in organisms and is commonly used as a plasticizer. Exposure to DEHP impairs the function of islet beta cells (INS-1 cells), which is related to insulin resistance and type 2 diabetes. At present, some research data have also confirmed that MEHP has a certain damage effect on INS-1 cells. In our experiment, we found that MEHP would lead to the increase of reactive oxygen species (ROS) and the upregulation of autophagy. And downregulated ROS production by N-acetyl-L-cysteine could also reduce autophagy. In addition, MEHP-induced lysosomal membrane permeability (LMP) subsequently released cathepsin D. Additionally, MEHP induced the collapse of mitochondrial transmembrane potential and release of cytochrome c. Addition of autophagy inhibitor 3-methyladenine relieved MEHP-induced apoptosis as assessed by the expression of cleaved caspase 3, cleaved caspase 9, and terminal deoxynucleotidyl transferase dUTP nick end labeling assay, indicating that MEHP-induced apoptosis was autophagy dependent. Cathepsin D inhibitor, pepstatin A, suppressed MEHP-induced mitochondria release of cytochrome c and apoptosis as well. Meanwhile, pyrroloquinoline quinone (PQQ), a new B vitamin, improved the above phenomenon. Taken together, our results indicate that MEHP induces autophagy-dependent apoptosis in INS-1 cells by lysosomal-mitochondrial axis. PQQ improved this process by downregulating ROS and provided a degree of protection. Our study provides a new perspective for MEHP on the cytotoxic mechanism and PQQ protection in INS-1 cells.


Assuntos
Apoptose/efeitos dos fármacos , Morte Celular Autofágica/efeitos dos fármacos , Dietilexilftalato/toxicidade , Neoplasias/tratamento farmacológico , Cofator PQQ/uso terapêutico , Animais , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais , Ratos
18.
Int J Biol Sci ; 15(1): 58-68, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30662347

RESUMO

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.


Assuntos
Estrogênios/deficiência , Osteoporose/etiologia , Osteoporose/prevenção & controle , Cofator PQQ/uso terapêutico , Animais , Senescência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Osteócitos/efeitos dos fármacos , Osteócitos/metabolismo , Osteoporose/metabolismo , Estresse Oxidativo/efeitos dos fármacos
19.
Neurosci Lett ; 687: 104-110, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30240821

RESUMO

Pyrroloquinoline quinone (PQQ), a redox cofactor in the mitochondrial respiratory chain, has been reported to protect SH-SY5Y cells from cytotoxicity induced by rotenone, a mitochondrial complex I inhibitor. In this study, we aimed to investigate the mitochondrial mechanisms involved in the neuroprotection of PQQ both in vitro and in vivo. The cultured human SH-SY5Y neuroblastoma cells were exposed to different concentrations of PQQ after which the cells were treated with rotenone. Electron microscopy images showed that PQQ could prevent the mitochondrial morphology damage. The down-regulation of mitochondrial biogenesis related genes (PGC-1alpha and TFAM) and mitochondrial fission and fusion related genes (Drp1and Mfn2) in rotenone-injured SH-SY5Y cells could be inhibited by PQQ. PQQ could also promote the transposition of Drp1 and Mfn2 from cytosol to mitochondria. In addition, rotenone was injected into the left medial forebrain bundle of SD rats to establish a Parkinson's disease (PD) model in vivo, after which different doses of PQQ or Edaravone were given intraperitoneally once daily for 8 weeks. PQQ could up-regulate the mRNA levels of PGC-1alpha, TFAM, Drp-1 and Mfn2 in the midbrain of PD rats. Our findings indicated that PQQ could prevent mitochondrial dysfunction by promoting mitochondrial biogenesis and regulating mitochondrial fission and fusion, which might contribute to its neuroprotective effect in PD models.


Assuntos
Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Cofator PQQ/uso terapêutico , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/prevenção & controle , Rotenona/toxicidade , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Inseticidas/toxicidade , Cofator PQQ/farmacologia , Transtornos Parkinsonianos/patologia , Ratos , Ratos Sprague-Dawley
20.
CNS Neurol Disord Drug Targets ; 16(6): 724-736, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28124619

RESUMO

BACKGROUND: Pyrroloquinoline quinone is an anionic, water-soluble compound with antioxidant characteristic. The role of pyrroloquinoline quinone in pharmacology and nutrition has attracted wide attention of researchers. Although a few experiments have confirmed that pyrroloquinoline quinone plays an obvious effective role in neuroprotection. There are few reports about the effect of pyrroloquinoline quinone on traumatic brain injury. Traumatic brain injury is one of the leading causes for adult disability and death. So far, there are no effective treatment methods for the injury because of its complex pathophysiology. METHOD: In the present study, a model of traumatic brain injury in rat was established to study the role of pyrroloquinoline quinone in central nervous system injury. RESULTS: The results showed that the protein expression of cleaved-Caspase 3/Caspase 3 increased after traumatic brain injury and the expression decreased by treatment with 2mM pyrroloquinoline quinone. Terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL) staining displayed that the TUNEL positive signals were up-regulated after traumatic brain injury and were down-regulated after treatment with 2mM pyrroloquinoline quinone. The protein expression of LC3II/LC3I or lysosome-associated membrane protein 2(LAMP2) was elevated after traumatic brain injury and reduced after administration with 2mM pyrroloquinoline quinone. Transmission electron microscopy showed that the number of autophagosomes increased markedly after traumatic brain injury and decreased on administration of 2mM pyrroloquinoline quinone. Electroencephalogram indicated that pyrroloquinoline quinone improved brain electrophysiological function after traumatic brain injury. The results of CCK-8 test showed that pyrroloquinoline quinone could increase the viability of primary astrocyte treated with Glutamate. Lactate dehydrogenase (LDH) assay demonstrated that pyrroloquinoline quinone decreased LDH content in primary astrocyte exposed to Glutamate. CONCLUSION: Pyrroloquinoline quinone could play a neuroprotective role after traumatic brain injury in rat, which might be associated with inhibiting apoptosis and autophagy caused by traumatic brain injury.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/patologia , Fármacos Neuroprotetores/uso terapêutico , Cofator PQQ/uso terapêutico , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Autofagossomos/efeitos dos fármacos , Autofagossomos/patologia , Autofagossomos/ultraestrutura , Lesões Encefálicas Traumáticas/fisiopatologia , Caspase 3/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Ácido Glutâmico/toxicidade , L-Lactato Desidrogenase/metabolismo , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Sprague-Dawley , Sincalida/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA