Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 118
1.
J Med Invest ; 71(1.2): 23-28, 2024.
Article En | MEDLINE | ID: mdl-38735721

Pyrroloquinoline quinone disodium salt (PQQ) is a red trihydrate crystal that was approved as a new food ingredient by FDA in 2008. Now, it is approved as a food in Japan and the EU. PQQ has redox properties and exerts antioxidant, neuroprotective, and mitochondrial biogenesis effects. The baseline intake level of PQQ is considered to be 20 mg/day. PQQ ingestion lowers blood lipid peroxide levels in humans, suggesting antioxidant activity. In the field of cognitive function, double-blind, placebo-controlled trials have been conducted. Various improvements have been reported regarding general memory, verbal memory, working memory, and attention. Furthermore, a stratified analysis of a population with a wide range of ages revealed unique effects in young people (20-40 years old) that were not observed in older adults (41-65 years old). Specifically, cognitive flexibility and executive speed improved more rapidly in young people at 8 weeks. Co-administration of PQQ and coenzyme Q10 further enhanced these effects. In an open-label trial, PQQ was shown to improve sleep and mood. Additionally, PQQ was found to suppress skin moisture loss and increase PGC-1α expression. Overall, PQQ is a food with various functions, including brain health benefits. J. Med. Invest. 71 : 23-28, February, 2024.


Brain , Cognition , PQQ Cofactor , Humans , PQQ Cofactor/pharmacology , PQQ Cofactor/administration & dosage , Cognition/drug effects , Brain/drug effects , Brain/metabolism , Antioxidants/pharmacology , Antioxidants/administration & dosage
2.
Stem Cell Res Ther ; 15(1): 97, 2024 Apr 05.
Article En | MEDLINE | ID: mdl-38581065

BACKGROUND: DNA damage and oxidative stress induced by chemotherapy are important factors in the onset of premature ovarian insufficiency (POI). Studies have shown that mitochondria derived from mesenchymal stem cells (MSC-Mito) are beneficial for age-related diseases, but their efficacy alone is limited. Pyrroloquinoline quinone (PQQ) is a potent antioxidant with significant antiaging and fertility enhancement effects. This study aimed to investigate the therapeutic effect of MSC-Mito in combination with PQQ on POI and the underlying mechanisms involved. METHODS: A POI animal model was established in C57BL/6J mice by cyclophosphamide and busulfan. The effects of MSC-Mito and PQQ administration on the estrous cycle, ovarian pathological damage, sex hormone secretion, and oxidative stress in mice were evaluated using methods such as vaginal smears and ELISAs. Western blotting and immunohistochemistry were used to assess the expression of SIRT1, PGC-1α, and ATM/p53 pathway proteins in ovarian tissues. A cell model was constructed using KGN cells treated with phosphoramide mustard to investigate DNA damage and apoptosis through comet assays and flow cytometry. SIRT1 siRNA was transfected into KGN cells to further explore the role of the SIRT1/ATM/p53 pathway in combination therapy with MSC-Mito and PQQ for POI. RESULTS: The combined treatment of MSC-Mito and PQQ significantly restored ovarian function and antioxidant capacity in mice with POI. This treatment also reduced the loss of follicles at various stages, improving the disrupted estrous cycle. In vitro experiments demonstrated that PQQ facilitated the proliferation of MitoTracker-labelled MSC-Mito, synergistically restoring mitochondrial function and inhibiting oxidative stress in combination with MSC-Mito. Both in vivo and in vitro, the combination of MSC-Mito and PQQ increased mitochondrial biogenesis mediated by SIRT1 and PGC-1α while inhibiting the activation of ATM and p53, consequently reducing DNA damage-mediated cell apoptosis. Furthermore, pretreatment of KGN cells with SIRT1 siRNA reversed nearly all the aforementioned changes induced by the combined treatment. CONCLUSIONS: Our research findings indicate that PQQ facilitates MSC-Mito proliferation and, in combination with MSC-Mito, ameliorates chemotherapy-induced POI through the SIRT1/ATM/p53 signaling pathway.


Mesenchymal Stem Cells , Primary Ovarian Insufficiency , Animals , Female , Humans , Mice , Antioxidants/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , PQQ Cofactor/pharmacology , Primary Ovarian Insufficiency/pathology , RNA, Small Interfering/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
J Cell Mol Med ; 28(8): e18299, 2024 Apr.
Article En | MEDLINE | ID: mdl-38613355

Pulmonary fibrosis is a lung disorder affecting the lungs that involves the overexpressed extracellular matrix, scarring and stiffening of tissue. The repair of lung tissue after injury relies heavily on Type II alveolar epithelial cells (AEII), and repeated damage to these cells is a crucial factor in the development of pulmonary fibrosis. Studies have demonstrated that chronic exposure to PM2.5, a form of air pollution, leads to an increase in the incidence and severity of pulmonary fibrosis by stimulation of epithelial-mesenchymal transition (EMT) in lung epithelial cells. Pyrroloquinoline quinone (PQQ) is a bioactive compound found naturally that exhibits potent anti-inflammatory and anti-oxidative properties. The mechanism by which PQQ prevents pulmonary fibrosis caused by exposure to PM2.5 through EMT has not been thoroughly discussed until now. In the current study, we discovered that PQQ successfully prevented PM2.5-induced pulmonary fibrosis by targeting EMT. The results indicated that PQQ was able to inhibit the expression of type I collagen, a well-known fibrosis marker, in AEII cells subjected to long-term PM2.5 exposure. We also found the alterations of cellular structure and EMT marker expression in AEII cells with PM2.5 incubation, which were reduced by PQQ treatment. Furthermore, prolonged exposure to PM2.5 considerably reduced cell migratory ability, but PQQ treatment helped in reducing it. In vivo animal experiments indicated that PQQ could reduce EMT markers and enhance pulmonary function. Overall, these results imply that PQQ might be useful in clinical settings to prevent pulmonary fibrosis.


Pulmonary Fibrosis , Animals , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , PQQ Cofactor/pharmacology , Epithelial-Mesenchymal Transition , Alveolar Epithelial Cells , Particulate Matter/toxicity
4.
Adv Sci (Weinh) ; 11(18): e2308970, 2024 May.
Article En | MEDLINE | ID: mdl-38454653

Alzheimer's disease (AD) is a pressing concern in neurodegenerative research. To address the challenges in AD drug development, especially those targeting Aß, this study uses deep learning and a pharmacological approach to elucidate the potential of pyrroloquinoline quinone (PQQ) as a neuroprotective agent for AD. Using deep learning for a comprehensive molecular dataset, blood-brain barrier (BBB) permeability is predicted and the anti-inflammatory and antioxidative properties of compounds are evaluated. PQQ, identified in the Mediterranean-DASH intervention for a diet that delays neurodegeneration, shows notable BBB permeability and low toxicity. In vivo tests conducted on an Aß1₋42-induced AD mouse model verify the effectiveness of PQQ in reducing cognitive deficits. PQQ modulates genes vital for synapse and anti-neuronal death, reduces reactive oxygen species production, and influences the SIRT1 and CREB pathways, suggesting key molecular mechanisms underlying its neuroprotective effects. This study can serve as a basis for future studies on integrating deep learning with pharmacological research and drug discovery.


Alzheimer Disease , Deep Learning , Disease Models, Animal , Neuroprotective Agents , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Neuroprotective Agents/pharmacology , Mice , PQQ Cofactor/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Male
5.
J Appl Toxicol ; 44(2): 235-244, 2024 Feb.
Article En | MEDLINE | ID: mdl-37650462

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.


Gentamicins , Ototoxicity , Humans , Gentamicins/metabolism , Reactive Oxygen Species/metabolism , PQQ Cofactor/pharmacology , PQQ Cofactor/therapeutic use , PQQ Cofactor/metabolism , Ototoxicity/etiology , Ototoxicity/prevention & control , Ototoxicity/metabolism , Hair Cells, Auditory/metabolism , Anti-Bacterial Agents/metabolism , Apoptosis
6.
Br J Nutr ; 131(8): 1352-1361, 2024 Apr 28.
Article En | MEDLINE | ID: mdl-38155410

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.


Dietary Supplements , Lipopolysaccharides , Animals , Swine , PQQ Cofactor/pharmacology , PQQ Cofactor/therapeutic use , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Liver/metabolism , RNA, Messenger/metabolism
7.
Fluids Barriers CNS ; 20(1): 84, 2023 Nov 20.
Article En | MEDLINE | ID: mdl-37981683

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.


Brain , PQQ Cofactor , Animals , Mice , PQQ Cofactor/pharmacology , PQQ Cofactor/therapeutic use , Reactive Oxygen Species , Folic Acid/pharmacology , DNA, Mitochondrial
8.
Cell Death Dis ; 14(11): 723, 2023 11 07.
Article En | MEDLINE | ID: mdl-37935689

Abnormal lipid metabolism and chronic low-grade inflammation are the main traits of obesity. Especially, the molecular mechanism of concomitant deficiency in steroidogenesis-associated enzymes related to testosterone (T) synthesis of obesity dominated a decline in male fertility is still poorly understood. Here, we found that in vivo, supplementation of pyrroloquinoline quinone (PQQ) efficaciously ameliorated the abnormal lipid metabolism and testicular spermatogenic function from high-fat-diet (HFD)-induced obese mice. Moreover, the transcriptome analysis of the liver and testicular showed that PQQ supplementation not only inhibited the high expression of proprotein convertase subtilisin/Kexin type 9 (PCSK9) but also weakened the NOD-like receptor family pyrin domain containing 3 (NLRP3)-mediated pyroptosis, which both played a negative role in T synthesis of Leydig Cells (LCs). Eventually, the function and the pyroptosis of LCs cultured with palmitic acid in vitro were simultaneously benefited by suppressing the expression of NLRP3 or PCSK9 respectively, as well the parallel effects of PQQ were affirmed. Collectively, our data revealed that PQQ supplementation is a feasible approach to protect T synthesis from PCSK9-NLRP3 crosstalk-induced LCs' pyroptosis in obese men.


NLR Family, Pyrin Domain-Containing 3 Protein , Proprotein Convertase 9 , Humans , Mice , Animals , Male , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , PQQ Cofactor/pharmacology , Mice, Obese , Leydig Cells/metabolism , Pyroptosis , Obesity/metabolism , Inflammation
9.
Acta Neuropathol Commun ; 11(1): 146, 2023 09 08.
Article En | MEDLINE | ID: mdl-37684640

Retinal ganglion cells are highly metabolically active requiring strictly regulated metabolism and functional mitochondria to keep ATP levels in physiological range. Imbalances in metabolism and mitochondrial mechanisms can be sufficient to induce a depletion of ATP, thus altering retinal ganglion cell viability and increasing cell susceptibility to death under stress. Altered metabolism and mitochondrial abnormalities have been demonstrated early in many optic neuropathies, including glaucoma, autosomal dominant optic atrophy, and Leber hereditary optic neuropathy. Pyrroloquinoline quinone (PQQ) is a quinone cofactor and is reported to have numerous effects on cellular and mitochondrial metabolism. However, the reported effects are highly context-dependent, indicating the need to study the mechanism of PQQ in specific systems. We investigated whether PQQ had a neuroprotective effect under different retinal ganglion cell stresses and assessed the effect of PQQ on metabolic and mitochondrial processes in cortical neuron and retinal ganglion cell specific contexts. We demonstrated that PQQ is neuroprotective in two models of retinal ganglion cell degeneration. We identified an increased ATP content in healthy retinal ganglion cell-related contexts both in in vitro and in vivo models. Although PQQ administration resulted in a moderate effect on mitochondrial biogenesis and content, a metabolic variation in non-diseased retinal ganglion cell-related tissues was identified after PQQ treatment. These results suggest the potential of PQQ as a novel neuroprotectant against retinal ganglion cell death.


Neuroprotection , Neuroprotective Agents , Retinal Ganglion Cells , PQQ Cofactor/pharmacology , Neuroprotective Agents/pharmacology , Adenosine Triphosphate
10.
Aesthet Surg J ; 44(1): NP104-NP118, 2023 Dec 14.
Article En | MEDLINE | ID: mdl-37616573

BACKGROUND: Reducing absorption after autologous fat grafting is a current challenge. Pyrroloquinoline quinone (PQQ) is the strongest known catalyst of redox reactions, which can scavenge reactive oxygen species (ROS) and alleviate oxidative stress. OBJECTIVES: The aim of this study was to establish an in vivo model of PQQ-assisted lipotransfer and clarify the role of PQQ in reducing oxidative stress, alleviating apoptosis, and promoting angiogenesis during the acute hypoxic phase after grafting. In addition the study was performed to assess whether this intervention would have a positive effect on the improvement of long-term volume retention. METHODS: Different concentrations of PQQ (low: 10 µM, medium: 100 µM, and high: 1000 µM) were mixed with human adipose tissue and transplanted subcutaneously into nude mice. Meanwhile, a control group of phosphate-buffered saline in an equal volume to PQQ was set up. On the third day after grafting, whole mount fluorescence staining was applied to detect ROS, mitochondrial membrane potential (MMP), apoptosis, adipocyte activity, and angiogenesis. Graft volume retention rate and electron microscopic morphology were evaluated at the third month. Immunohistochemistry and polymerase chain reaction (PCR) were further employed to elucidate the mechanism of action of PQQ. RESULTS: PQQ-assisted fat grafting improved the long-term volume retention, promoted the quality and viability of the adipose tissue, and reduced the level of fibrosis. The underlying mechanism of PQQ assisted in scavenging the accumulated ROS, restoring MMP, enhancing adipocyte viability, alleviating tissue apoptosis, and promoting timely angiogenesis during the hypoxia stress phase. The most effective concentration of PQQ was 100 µM. Immunohistochemistry and PCR experiments confirmed that PQQ reduced the expression of Bax and cytochrome c in the mitochondrial apoptotic pathway and increased the level of the antiapoptotic molecule Bcl-2. CONCLUSIONS: PQQ could improve the long-term survival of adipocytes by alleviating hypoxic stress and promoting timely angiogenesis in the early phase following lipotransfer.


Angiogenesis , PQQ Cofactor , Mice , Animals , Humans , Reactive Oxygen Species/metabolism , PQQ Cofactor/pharmacology , PQQ Cofactor/metabolism , Mice, Nude , Oxidative Stress
11.
Cell Mol Biol (Noisy-le-grand) ; 69(4): 60-69, 2023 Apr 30.
Article En | MEDLINE | ID: mdl-37329547

Our study aimed to reveal the effects and changes, antioxidant metabolism (Oxidative Stress), inflammatory response, mitochondrial biogenesis and mitochondrial dysfunction characteristics in hepatocellular carcinoma cell line HepG2; that occur in genes (NRF-1, NRF-2, NFκB and PGC-1α) and miRNAs (miR15-a, miR16-1, miR181-c) that can control related features. To investigate the effects of Pyrroloquinoline quinone (PQQ) and Coenzyme Q10 (CoQ10) in HepG2, and their effects on cell viability, lateral cell migration, gene expression and miRNA expression levels were investigated. If the data we have obtained are evaluated in terms of anti-cancer effectiveness, the most effective use of CoQ10 can be defined as the use alone rather than the combined use. According to the results of the wound healing experiment, we determined that Pyrroloquinoline quinone and combined drug application increased the wound closure area and cell proliferation compared to the control group, while CoQ10 application decreased it. We found that Pyrroloquinoline quinone and Coenzyme Q10 exposure in the HepG2 cell line increased Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) expression but not NRF-1 gene expression. We reported only a small increase in expression of the NRF-2 gene in the Pyrroloquinoline quinone application compared to the control group. We found that only Pyrroloquinoline quinone and CoQ10 application caused more expression increase in the Nuclear Factor kappa B (NFκB) gene compared to combined application. Pyrroloquinoline quinone and CoQ10 administration down-regulated the expression levels of miR16-1, miR15a and miR181c. The use of Pyrroloquinoline quinone and CoQ10 is effective on epigenetic factors, miR-15a, miR-16-1 and miR181c are important candidate biomarkers in hepatocellular carcinoma and diseases accompanied by mitochondrial dysfunction.


Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Humans , Transcription Factors/genetics , PQQ Cofactor/pharmacology , PQQ Cofactor/genetics , PQQ Cofactor/metabolism , Mitochondria , Genes, Mitochondrial , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Cell Line
12.
Aging Cell ; 22(9): e13912, 2023 09.
Article En | MEDLINE | ID: mdl-37365714

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.


Bone Resorption , Osteoporosis , Mice , Animals , Antioxidants/metabolism , PQQ Cofactor/pharmacology , PQQ Cofactor/metabolism , PQQ Cofactor/therapeutic use , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Up-Regulation , Fibrillin-1/metabolism , Molecular Docking Simulation , Oxidative Stress , Aging , Osteoporosis/drug therapy , Osteoporosis/metabolism , Bone Resorption/drug therapy
13.
J Dev Orig Health Dis ; 14(3): 321-324, 2023 06.
Article En | MEDLINE | ID: mdl-36861270

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.


Fetal Growth Retardation , Myocytes, Cardiac , Animals , Female , Guinea Pigs , Pregnancy , Antioxidants , Fetal Growth Retardation/drug therapy , Fetal Growth Retardation/metabolism , Oxidation-Reduction , PQQ Cofactor/pharmacology , PQQ Cofactor/therapeutic use
14.
Mediators Inflamm ; 2022: 1267841, 2022.
Article En | MEDLINE | ID: mdl-36345503

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.


Asthma , PQQ Cofactor , Animals , Mice , Asthma/metabolism , Bronchoalveolar Lavage Fluid , Cytokines/metabolism , Disease Models, Animal , Inflammation/metabolism , Lung/metabolism , Mice, Inbred BALB C , Ovalbumin , PQQ Cofactor/pharmacology , PQQ Cofactor/therapeutic use , PQQ Cofactor/metabolism , Signal Transduction , Th2 Cells , STAT Transcription Factors/metabolism , Janus Kinases
15.
Sci Rep ; 12(1): 15911, 2022 09 23.
Article En | MEDLINE | ID: mdl-36151123

We investigated whether the oxidoreductase cofactor pyrroloquinoline quinone (PQQ) prevents noise-induced and age-related hearing loss (NIHL and ARHL) in mice. To assess NIHL, 8 week-old mice with and without PQQ administration were exposed to noise for 4 h. PQQ was orally administered for one week before and after noise exposure and subcutaneously once before noise exposure. For ARHL evaluation, mice were given drinking water with or without PQQ starting at 2 months of age. In the NIHL model, PQQ-treated mice had auditory brainstem response (ABR) thresholds of significantly reduced elevation at 8 kHz, a significantly increased number of hair cells at the basal turn, and significantly better maintained synapses beneath the inner hair cells compared to controls. In the ARHL model, PQQ significantly attenuated the age-related increase in ABR thresholds at 8 and 32 kHz at 10 months of age compared to controls. In addition, the hair cells, spiral ganglion cells, ribbon synapses, stria vascularis and nerve fibers were all significantly better maintained in PQQ-treated animals compared to controls at 10 months of age. These physiological and histological results demonstrate that PQQ protects the auditory system from NIHL and ARHL in mice.


Hearing Loss, Noise-Induced , PQQ Cofactor , Presbycusis , Aging , Animals , Auditory Threshold/physiology , Cochlea/pathology , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing Loss, Noise-Induced/pathology , Mice , Oxidoreductases , PQQ Cofactor/pharmacology , Presbycusis/pathology
16.
Pulm Pharmacol Ther ; 76: 102156, 2022 10.
Article En | MEDLINE | ID: mdl-36030026

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.


Hypertension, Pulmonary , Animals , Endothelial Cells , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Inflammation/drug therapy , Monocrotaline , PQQ Cofactor/metabolism , PQQ Cofactor/pharmacology , PQQ Cofactor/therapeutic use , Pulmonary Artery , Rats , Rats, Sprague-Dawley
17.
Int J Mol Sci ; 23(11)2022 May 27.
Article En | MEDLINE | ID: mdl-35682720

Maternal obesity and consumption of a high-fat diet significantly elevate risk for pediatric nonalcoholic fatty liver disease (NAFLD), affecting 10% of children in the US. Almost half of these children are diagnosed with nonalcoholic steatohepatitis (NASH), a leading etiology for liver transplant. Animal models show that signs of liver injury and perturbed lipid metabolism associated with NAFLD begin in utero; however, safe dietary therapeutics to blunt developmental programming of NAFLD are unavailable. Using a mouse model of maternal Western-style diet (WD), we previously showed that pyrroloquinoline quinone (PQQ), a potent dietary antioxidant, protected offspring of WD-fed dams from development of NAFLD and NASH. Here, we used untargeted mass spectrometry-based lipidomics to delineate lipotoxic effects of WD on offspring liver and identify lipid targets of PQQ. PQQ exposure during pregnancy altered hepatic lipid profiles of WD-exposed offspring, upregulating peroxisome proliferator-activated receptor (PPAR) α signaling and mitochondrial fatty acid oxidation to markedly attenuate triglyceride accumulation beginning in utero. Surprisingly, the abundance of very long-chain ceramides, important in promoting gut barrier and hepatic function, was significantly elevated in PQQ-treated offspring. PQQ exposure reduced the hepatic phosphatidylcholine/phosphatidylethanolamine (PC/PE) ratio in WD-fed offspring and improved glucose tolerance. Notably, levels of protective n - 3 polyunsaturated fatty acids (PUFAs) were elevated in offspring exposed to PQQ, beginning in utero, and the increase in n - 3 PUFAs persisted into adulthood. Our findings suggest that PQQ supplementation during gestation and lactation augments pathways involved in the biosynthesis of long-chain fatty acids and plays a unique role in modifying specific bioactive lipid species critical for protection against NAFLD risk in later life.


Fatty Acids, Omega-3 , Non-alcoholic Fatty Liver Disease , Adult , Animals , Child , Diet, High-Fat/adverse effects , Dietary Supplements , Fatty Acids, Omega-3/metabolism , Female , Humans , Lipid Metabolism , Liver/metabolism , Longevity , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress , PPAR alpha/metabolism , PQQ Cofactor/pharmacology , Pregnancy
18.
ACS Chem Neurosci ; 13(8): 1178-1186, 2022 04 20.
Article En | MEDLINE | ID: mdl-35413176

Parkinson's disease (PD) is associated with the aggregation and misfolding of a-synuclein (a-syn) protein in dopaminergic neurons. The misfolding process is heavily linked to copper dysregulation in PD. Experimental evidence supports the hypothesis that the co-presence of Cu(II) and α-syn facilitates the aggregation of α-syn, affecting the pathological development of PD. Recent literature has shown that pyrroloquinoline quinone (PQQ) contains strong neuroprotective activity by reducing the reactive oxygen species (ROS) production by α-syn. Despite these known facts, minimal studies have been done on the antioxidant effect of PQQ against ROS formation in the presence of Cu(II) and α-syn-119. Thus, it is of great significance to study the interaction between all three components, PQQ, Cu(II), and α-syn-119. In this proof-of-concept study, a variety of chemical techniques were employed to examine the antioxidant effect of PQQ on ROS that α-syn-119 produced in the presence of Cu(II). Our results showed that PQQ effectively prevented ROS formation in SH-SY5Y human differentiated neuronal cells. Thioflavin T (ThT) fluorescence assay, circular dichroism (CD) spectroscopy, and transmission electron microscopy (TEM) were applied, where PQQ was able to actively prevent fibrillation of α-syn-119 in the presence of Cu(II). This finding was further confirmed using electrochemical impedance spectroscopy (EIS), where the binding of PQQ to the α-syn-119 suppressed the aggregation process on the electrode surface. With these encouraging results, we envisage that PQQ and its derivatives can be a promising candidate for further studies as a multitarget therapeutic agent toward PD therapy.


Parkinson Disease , alpha-Synuclein , Antioxidants/pharmacology , Copper , Dopaminergic Neurons/metabolism , Humans , PQQ Cofactor/pharmacology , Parkinson Disease/drug therapy , Reactive Oxygen Species/metabolism , alpha-Synuclein/metabolism
19.
Biomed Pharmacother ; 150: 112998, 2022 Jun.
Article En | MEDLINE | ID: mdl-35489281

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.


Diabetes Mellitus, Experimental , Diabetic Nephropathies , Animals , Caspase 1 , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Fibrosis , Glucose/pharmacology , Humans , Interleukin-18 , Kidney , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , PQQ Cofactor/pharmacology , PQQ Cofactor/therapeutic use , Pyroptosis , Reactive Oxygen Species/metabolism
20.
Poult Sci ; 101(5): 101812, 2022 May.
Article En | MEDLINE | ID: mdl-35325831

As the antioxidant capacity of sperm declines with age in roosters, the objective of the present study was to determine the effects of different levels of pyrroloquinoline quinone disodium (PQQ.Na2) on antioxidative and sperm quality parameters of aging layer breeder roosters. A total of ninety-six 63-wk-old Jinghong No. 1 layer breeder roosters were randomly assigned to 4 treatments (0, 0.5, 1, 2 mg/kg PQQ.Na2) for 6 wk. Antioxidant activity and semen parameters were assessed biweekly. The dietary administration of PQQ.Na2 significantly increased semen quality (semen volume, sperm motility, straightness, progressive motility, curvilinear velocity, straight-line velocity, and amplitude of lateral head displacement) and antioxidant capacity (T-SOD, GSH-Px, hydroxyl radical scavenging ability, and/or superoxide scavenging capacity) in seminal plasma in aging layer breeder roosters. Whereas, PQQ.Na2 supplementations significantly decreased malondialdehyde (MDA) concentration in seminal plasma in aging layer breeder roosters. Supplementation with 1 mg/kg dietary PQQ.Na2 as an antioxidant supplement could increase sperm quality and antioxidant activity of aging layer breeder roosters, while a higher dose (2 mg/kg) did not result in further increment.


PQQ Cofactor , Semen Analysis , Aging , Animal Feed/analysis , Animals , Antioxidants/metabolism , Chickens/metabolism , Dietary Supplements , Male , Oxidative Stress , PQQ Cofactor/pharmacology , Semen , Semen Analysis/veterinary , Sperm Motility
...