Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
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.
ISME J ; 17(11): 2103-2111, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37737251

RESUMO

Although microbial humus respiration plays a critical role in organic matter decomposition and biogeochemical cycling of elements in diverse anoxic environments, the role of methane-producing species (methanogens) is not well defined. Here we report that a major fraction of humus, humic acid reduction enhanced the growth of Methanosarcina acetivorans above that attributed to methanogenesis when utilizing the energy sources methanol or acetate, results which showed both respiratory and fermentative modes of energy conservation. Growth characteristics with methanol were the same for an identically cultured mutant deleted for the gene encoding a multi-heme cytochrome c (MmcA), results indicating MmcA is not essential for respiratory electron transport to humic acid. Transcriptomic analyses revealed that growth with humic acid promoted the upregulation of genes annotated as cell surface pyrroloquinoline quinone (PQQ)-binding proteins. Furthermore, PQQ isolated from the membrane fraction was more abundant in humic acid-respiring cells, and the addition of PQQ improved efficiency of the extracellular electron transport. Given that the PQQ-binding proteins are widely distributed in methanogens, the findings extend current understanding of microbial humus respiration in the context of global methane dynamics.


Assuntos
Substâncias Húmicas , Metanol , Metanol/metabolismo , Cofator PQQ/metabolismo , Methanosarcina/metabolismo , Metano/metabolismo
3.
Aesthet Surg J ; 44(1): NP104-NP118, 2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-37616573

RESUMO

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.


Assuntos
Angiogênese , Cofator PQQ , Camundongos , Animais , Humanos , Espécies Reativas de Oxigênio/metabolismo , Cofator PQQ/farmacologia , Cofator PQQ/metabolismo , Camundongos Nus , Estresse Oxidativo
4.
Cell Mol Biol (Noisy-le-grand) ; 69(4): 60-69, 2023 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-37329547

RESUMO

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.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroRNAs , Humanos , Fatores de Transcrição/genética , Cofator PQQ/farmacologia , Cofator PQQ/genética , Cofator PQQ/metabolismo , Mitocôndrias , Genes Mitocondriais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , MicroRNAs/metabolismo , Linhagem Celular
5.
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
6.
Angew Chem Int Ed Engl ; 62(18): e202216962, 2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-36637456

RESUMO

Poly(vinyl alcohol) (PVA) is a water-soluble synthetic vinyl polymer with remarkable physical properties including thermostability and viscosity. Its biodegradability, however, is low even though a large amount of PVA is released into the environment. Established physical-chemical degradation methods for PVA have several disadvantages such as high price, low efficiency, and secondary pollution. Biodegradation of PVA by microorganisms is slow and frequently involves pyrroloquinoline quinone (PQQ)-dependent enzymes, making it expensive due to the costly cofactor and hence unattractive for industrial applications. In this study, we present a modified PVA film with improved properties as well as a PQQ-independent novel enzymatic cascade for the degradation of modified and unmodified PVA. The cascade consists of four steps catalyzed by three enzymes with in situ cofactor recycling technology making this cascade suitable for industrial applications.


Assuntos
Cofator PQQ , Álcool de Polivinil , Álcool de Polivinil/química , Biodegradação Ambiental , Cofator PQQ/metabolismo
7.
Lab Med ; 54(1): 72-74, 2023 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-35976923

RESUMO

OBJECTIVE: Assays based on redox reactions that involve proton transfer are vulnerable to artifactual findings in metabolic acidosis/alkalosis. We evaluated the impact of pH on the measurement of blood glucose by the glucose dehydrogenase/pyrroloquinoline quinone system used in point-of-care-testing. METHODS: We applied a series of thermodynamic equations to adjust the Gibbs energy for the pyrroloquinoline quinone couple. This adjusts values taken under standard conditions to those more closely resembling the physiological state. RESULTS: Under standard conditions, the pyrroloquinoline quinone couple has Eo = -0.125 V whereas adjustment to the physiological state (pH 7.40, ionic strength 0.15 mol/L, and temperature 310.15°K) yields Eo' = -0.166 V. This corresponds to an uncertainty in blood glucose determination of approximately 0.13 mmol/L. CONCLUSION: We have demonstrated that the impact of pH on blood glucose determination by the glucose dehydrogenase/pyrroloquinoline quinone system (under physiologically relevant conditions of ionic strength and temperature) is not clinically significant.


Assuntos
Glicemia , Cofator PQQ , Humanos , Cofator PQQ/metabolismo , Glucose Desidrogenase/metabolismo , Oxirredução
8.
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
9.
J Oleo Sci ; 71(12): 1761-1767, 2022 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-36336345

RESUMO

Following a growing interest in the physiological effects of pyrroloquinoline quinone (PQQ), more cell culture experiments have begun to elucidate its mechanism of action. However, to our knowledge, no reports have used instrumental analysis, such as liquid chromatography-tandem mass spectrometry (LC-MS/MS), to study cellular uptake of PQQ. In addition, despite the propensity of PQQ to react with amino acids and other compounds, only a handful of cell culture experiments have been conducted on PQQ derivatives. In the present study, we prepared PQQ derivatives by reacting PQQ with various amino acids and used them as reference standards for optimizing the LC-MS/MS analysis conditions to detect PQQ and its derivatives. Using this method, we evaluated the uptake of PQQ into mouse 3T3-L1 cells and found that most PQQ added to the medium was taken up by the cells in its unchanged form, while some PQQ reacted with amino acids in the medium and was taken up by the cells as PQQ derivatives. These results suggest that PQQ derivatives may contribute to the physiological effects of PQQ. To further elucidate the function of PQQ, it is necessary for future studies to clarify the activity of PQQ derivatives and to evaluate the types of PQQ present in food, animal, and cell samples in more detail.


Assuntos
Cofator PQQ , Espectrometria de Massas em Tandem , Camundongos , Animais , Cofator PQQ/química , Cofator PQQ/metabolismo , Células 3T3-L1 , Cromatografia Líquida , Aminoácidos , Técnicas de Cultura de Células
10.
Appl Microbiol Biotechnol ; 106(23): 7751-7761, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36271931

RESUMO

5-Ketogluconate (5KGA) is a precursor for synthesizing tartrate, a valuable compound used in several industries. In a previous study, Gluconobacter japonicus NBRC 3271 mutant strain D2, which lacks two membranous gluconate 2-dehydrogenases, was shown to produce 5KGA but not 2-ketogluconate from a mixture of glucose and gluconate. In this study, we aimed to develop an efficient 5KGA production system using G. japonicus D2 as the parental strain. D2 produced 5KGA from glucose in a jar fermentor culture; however, 5KGA levels were reduced during the late phase of cultivation. To increase the potential of D2 for 5KGA production, the cytoplasmic metabolism related to the utilization of 5KGA and gluconate was modified; the gno and gntK genes encoding 5KGA reductase and gluconokinase, respectively, were deleted from D2, generating D4. Improved 5KGA production was observed in D4 compared to that in D2, but a significant amount of gluconate remained at the end of cultivation, leading to an unsatisfied yield of 0.83 mol (mol glucose)-1. The conversion of gluconate to 5KGA is catalyzed by pyrroloquinoline quinone (PQQ)-dependent glycerol dehydrogenase (GLDH), which easily forms an apoenzyme by releasing PQQ and calcium ions. Thus, the effects of CaCl2 addition to the culture medium on 5KGA production by D4 were investigated. We demonstrated that 1 mM CaCl2 addition positively affected the maintenance of the PQQ-GLDH activity toward gluconate and consequently enhanced 5KGA production, and the yield reached 0.97 mol (mol glucose)-1. KEY POINTS: • An efficient 5KGA production system was developed with Gluconobacter japonicus. • Deleting the gno and gntK genes blocked the catabolism of 5KGA and gluconate. • The addition of 1 mM CaCl2 efficiently improved the conversion of glucose to 5KGA.


Assuntos
Gluconobacter , Cloreto de Cálcio , Gluconatos/metabolismo , Cofator PQQ/metabolismo , Glucose/metabolismo
11.
Mol Microbiol ; 118(4): 417-425, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36054785

RESUMO

Glucose is taken up by Escherichia coli through the phosphotransferase system (PTS) as the preferred carbon source. PTS mutants grow with glucose as a carbon source only in the presence of pyrroloquinoline quinone (PQQ), which is needed as a redox cofactor for the glucose dehydrogenase Gcd. The membrane-anchored Gcd enzyme oxidises glucose to gluconolactone in the periplasm. For this reaction to occur, external supply of PQQ is required as E. coli is unable to produce PQQ de novo. Growth experiments show that PqqU (previously YncD) is the TonB-ExbBD-dependent transporter for PQQ through the outer membrane. PQQ protected the cells from the PqqU-dependent phage IsaakIselin (Bas10) by competition for the receptor protein. As a high affinity uptake system, PqqU allows E. coli to activate Gcd even at surrounding PQQ concentrations of about 1 nmoL/L. At about 30-fold higher PQQ concentrations, the activation of Gcd gets PqqU independent. Due to its small size, Pqq may also pass the outer membrane through porins. The PQQ-dependent production of gluconate has been demonstrated in many plant growth-promoting bacteria that solubilise phosphate minerals in the soil by secreting this acid. Under phosphate limiting conditions also E. coli induces the glucose dehydrogenase and secretes gluconate, even in absence of PTS, that is, even when the bacterium is unable to grow on glucose without PQQ.


Assuntos
Escherichia coli K12 , Cofator PQQ , Carbono/metabolismo , Escherichia coli/metabolismo , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Gluconatos/metabolismo , Glucose/metabolismo , Glucose 1-Desidrogenase/genética , Glucose 1-Desidrogenase/metabolismo , Fosfatos/metabolismo , Fosfotransferases/metabolismo , Porinas/metabolismo , Cofator PQQ/metabolismo , Quinonas/metabolismo , Solo
12.
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
13.
Microbiol Spectr ; 10(4): e0093322, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-35852313

RESUMO

Imbalances of soil available nutrients and soilborne diseases have seriously restricted the productivity of crops and jeopardized food security worldwide. Pyrroloquinoline quinone (PQQ), a redox cofactor in some bacteria involved in glucose metabolism and phosphorus mineralization, could be anticipated to alter soil ecosystems to a certain extent. However, there is limited information on PQQ defending soilborne pathogens and regulating soil main nutrients. Here, a pot experiment based on mono-cropping soils of pepper was conducted to examine the effects of PQQ amendment on reconstructing soil microbial communities and soil nutrients under aerobic/anaerobic conditions comprising three treatments, namely, control, PQQ (aerobic), and FL-PQQ (anaerobic). The results revealed that soil microbial community composition and soil nutrients were distinctly altered by PQQ regimes. Compared to control, PQQ treatment significantly increased the content of soil available phosphorus (AP), while FL_PQQ treatment strongly improved the content of soil available nitrogen (AN). In terms of pathogens, relative to control, both PQQ treatments suppressed the abundances of pathogens, of which FL_PQQ treatment significantly decreased the abundance of the pathotrophic fungal by 64% and the abundance of Fusarium oxysporum by 57%, largely attributed to the increase of organic acid generators (Oxobacter, Hydrogenispora) and potential antagonists (Bacillus, Talaromyces). Structural equation modeling (SEM) showed that PQQ regimes suppressed pathogens by indirectly regulating soil physicochemical properties and microbial communities. Overall, we proposed that PQQ application both in aerobic/anaerobic conditions could improve soil available nutrients and suppress soil pathogens in pepper monocropping soils. IMPORTANCE The attention to PQQ (pyrroloquinoline quinone) effect on soil nutrients and pathogens was less paid in monocropping soils. However, the underlying microbial interacting mechanism remains unclear. Adopting a novel external bio-additive, the effects of PQQ on soil main nutrients and the pathotrophic fungal under aerobic and anaerobic regimes will be investigated, which would help to improve soil quality health. Our main conclusion was that PQQ would help to remediate monocropping obstacle soils in terms of soil nutrients and soil pathogens by associating with the microbial community, and anaerobic PQQ application more favored amelioration of continuous obstacle soils. These results will benefit the health and sustainable development of pepper production as well as other greenhouse vegetable production.


Assuntos
Microbiota , Solo , Anaerobiose , Nutrientes , Cofator PQQ/química , Cofator PQQ/metabolismo , Fósforo , Solo/química
14.
J Agric Food Chem ; 70(22): 6764-6774, 2022 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-35613468

RESUMO

The pyrroloquinoline quinone (PQQ)-dependent dehydrogenase DepA detoxifies deoxynivalenol (DON) by converting the C3-OH into a keto group. Herein, two crystal structures of DepA and its complex with PQQ were determined, together with biochemical evidence confirming the interactions of DepA with PQQ and DON and revealing a unique tyrosine residue important for substrate selection. Furthermore, four loops over the active site essential for DepA activity were identified, of which three loops were stabilized by PQQ, and the fourth loop invisible in both structures was considered important for binding DON, together constituting a lid for the active site. Preliminary engineering of the loop showed its potential for enzyme improvement. This study provides structural insights into how a PQQ-dependent dehydrogenase is equipped with the function of DON conversion and for the first time shows the necessity of a lid structure for PQQ-dependent dehydrogenase activity, laying foundation for structure-based design to enhance catalysis efficiency.


Assuntos
Quinona Redutases , Tricotecenos , Cofator PQQ/química , Cofator PQQ/metabolismo , Quinonas , Tricotecenos/metabolismo
15.
Inorg Chem ; 61(15): 5943-5956, 2022 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-35362953

RESUMO

PqqB from Methylobacterium extorquens is a unique nonheme iron-dependent hydroxylase involved in the biosynthesis of redox cofactor pyrroloquinoline quinone (PQQ). A series of recent experiments have demonstrated that PqqB catalyzes the stepwise insertions of two oxygen atoms into the tyrosine ring of the diamino acid substrate, generating the quinone moiety of PQQ; however, the reaction details have not been elucidated yet. In this paper, on the basis of the crystal structures, the enzyme-substrate complex models were constructed, and the catalytic mechanism of PqqB was explored by performing a series of combined QM/MM calculations. Our results confirmed that the first hydroxylation is performed by the highly reactive FeIV-oxo species and follows the typical H-abstraction/hydroxyl rebound mechanism, which is similar to the common aliphatic hydroxylation catalyzed by the α-KG enzymes. Nevertheless, the second hydroxylation is achieved by the Fe-O2 species, and the reactant complex can be described as an intermediate radical-FeII-superoxide, that is, the dioxygen is activated by accepting an electron from the bidentate coordination intermediate. Since both the dioxygen and intermediate are activated by electron transfer, the distal oxygen of superoxide can directly attack the carbonyl carbon of substrate to form an alkylperoxo intermediate, then the O-O heterolysis occurs to afford the epoxide intermediate, which finally evolves into the product by rearrangement. It is the bidentate coordination of catechol moiety to iron that leads to the one-electron oxidation of the substrate by the dioxygen, which significantly activates the substrate and promotes the superoxide radical attack. During the catalysis, Asp90 and His240 in the second sphere play an important role by acting as acid-base catalysts to mediate the proton transfer and manipulate the suitable orientation of superoxide. These findings may provide useful information for understanding the unique reaction mechanism of PqqB that employs both the FeIV-oxo and FeII-superoxide to carry out the aromatic hydroxylation.


Assuntos
Oxigenases de Função Mista , Cofator PQQ , Catálise , Compostos Ferrosos , Hidroxilação , Ferro , Oxirredução , Oxigênio , Cofator PQQ/metabolismo , Superóxidos
16.
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
17.
Chem Res Toxicol ; 35(3): 355-377, 2022 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-35166521

RESUMO

The widely distributed, essential redox factor pyrroloquinoline quinone (PQQ, methoxatin) (1) was discovered in the mid-1960s. The breadth and depth of its biological effects are steadily being revealed, and understanding its biosynthesis at the genomic level is a continuing process. In this review, aspects of the chemistry, biology, biosynthesis, and commercial production of 1 at the gene level, and some applications, are presented from discovery through to mid-2021.


Assuntos
Biologia , Cofator PQQ , Oxirredução , Cofator PQQ/genética , Cofator PQQ/metabolismo
18.
J Bacteriol ; 204(3): e0055821, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35072518

RESUMO

Acetic acid bacteria catalyze the two-step oxidation of ethanol to acetic acid using the membrane-bound enzymes pyrroloquinoline quinone-dependent alcohol dehydrogenase and molybdopterin-dependent aldehyde dehydrogenase (ALDH). Although the reducing equivalents from the substrate are transferred to ubiquinone in the membrane, intramolecular electron transport in ALDH is not understood. Here, we purified the AldFGH complex, the membrane-bound ALDH that is physiologically relevant to acetic acid fermentation in Gluconacetobacter diazotrophicus strain PAL5. The purified AldFGH complex showed acetaldehyde:ubiquinone (Q2) oxidoreductase activity. c-type cytochromes of the AldFGH complex (in the AldF subunit) were reduced by acetaldehyde. Next, we genetically dissected the AldFGH complex into AldGH and AldF units and reconstituted them. The AldGH subcomplex showed acetaldehyde:ferricyanide oxidoreductase activity but not Q2 reductase activity. The ALDH activity of AldGH was not found in membranes but was found in the soluble fraction of the recombinant strain, suggesting that the AldF subunit is responsible for membrane binding of the AldFGH complex. The absorption spectra of the purified AldGH subcomplex suggested the presence of an [Fe-S] cluster, which can be reduced by acetaldehyde. The AldFGH complex reconstituted from the AldGH subcomplex and AldF showed Q2 reductase activity. We propose a model in which electrons from the substrate are abstracted by a molybdopterin in the AldH subunit and transferred to the [Fe-S] cluster(s) in the AldG subunit, followed by electron transport to c-type cytochrome centers in the AldF subunit, which is the site of ubiquinone reduction in the membrane. IMPORTANCE Two membrane-bound enzymes of acetic acid bacteria, pyrroloquinoline quinone-dependent alcohol dehydrogenase and molybdopterin-dependent aldehyde dehydrogenase (ALDH), are responsible for vinegar production. Upon the oxidation of acetaldehyde, ALDH reduces ubiquinone in the cytoplasmic membrane. ALDH is an enzyme complex of three subunits. Here, we tried to understand how ALDH works by using a classical biochemical approach and genetic engineering to dissect the enzyme complex into soluble and membrane-bound parts. The soluble part had limited activity in vitro and did not reduce ubiquinone. However, the enzyme complex reconstituted from the soluble and membrane-bound parts showed ubiquinone reduction activity. The proposed working model of ALDH provides a better understanding of how the enzyme works in the vinegar fermentation process.


Assuntos
Álcool Desidrogenase , Aldeído Desidrogenase , Acetaldeído , Ácido Acético/metabolismo , Álcool Desidrogenase/metabolismo , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Aldeídos , Citocromos/metabolismo , Transporte de Elétrons , Gluconacetobacter , Cofator PQQ/metabolismo , Ubiquinona/metabolismo
19.
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
20.
Environ Sci Pollut Res Int ; 29(20): 30383-30393, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34997497

RESUMO

The current study aimed to investigate the potential ameliorative effects of pyrroloquinoline quinone (PQQ) on cyclophosphamide (CTX)-induced liver injury in mice. The liver injury model was established by injecting mice with CTX (80 mg/kg/day). Liver function indices, antioxidant enzyme activities, and inflammatory cytokines were evaluated. In addition, protein expression levels of the nuclear factor E2-related factor 2 (Nrf2) and nuclear factor kappa-B (NF-κB) pathways in the liver tissues were determined using western blot. The results indicated that PQQ decreased the serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), and the malondialdehyde (MDA), interleukin (IL)-1ß, IL-6, tumor necrosis factor-α (TNF-α) levels in the liver tissues. Moreover, PQQ enhanced the activities of oxidative stress markers to alleviate CTX induced oxidative stress. Furthermore, the expression levels of heme oxygenase-1 (HO-1), glutamate-cysteine ligase modifier subunit (GCLM), and NAD(P)H quinone oxidoreductase 1 (NQO1) were significantly increased, and the expression levels of NF-κB p50, NF-κB p65, and inhibitor of NF-κB kinase alpha (IKKα) were significantly decreased after PQQ administration, suggesting that PQQ alleviated CTX-induced liver injury via activating the Nrf2-mediated antioxidant response pathway, and inhibiting the NF-κB-mediated inflammation pathway. Therefore, PQQ can be potentially used as a dietary supplement or functional foods for alleviating the CTX-induced liver injury.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , Fator 2 Relacionado a NF-E2 , Animais , Antioxidantes/metabolismo , Doença Hepática Crônica Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Crônica Induzida por Substâncias e Drogas/patologia , Ciclofosfamida/metabolismo , Ciclofosfamida/toxicidade , Fígado , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo , Cofator PQQ/metabolismo , Cofator PQQ/farmacologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...