Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Mol Biosci ; 11: 1386598, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38721278

RESUMO

Humans interact with a multitude of microorganisms in various ecological relationships, ranging from commensalism to pathogenicity. The same applies to fungi, long recognized for their pathogenic roles in infection-such as in invasive fungal diseases caused, among others, by Aspergillus fumigatus and Candida spp.-and, more recently, for their beneficial activities as an integral part of the microbiota. Indeed, alterations in the fungal component of the microbiota, or mycobiota, have been associated with inflammatory, infectious and metabolic diseases, and cancer. Whether acting as opportunistic pathogens or symbiotic commensals, fungi possess a complex enzymatic repertoire that intertwines with that of the host. In this metabolic cross-talk, fungal enzymes may be unique, thus providing novel metabolic opportunities to the host, or, conversely, produce toxic metabolites. Indeed, administration of fungal probiotics and fungi-derived products may be beneficial in inflammatory and infectious diseases, but fungi may also produce a plethora of toxic secondary metabolites, collectively known as mycotoxins. Fungal enzymes may also be homologues to human enzymes, but nevertheless embedded in fungal-specific metabolic networks, determined by all the interconnected enzymes and molecules, quantitatively and qualitatively specific to the network, such that the activity and metabolic effects of each enzyme remain unique to fungi. In this Opinion, we explore the concept that targeting this fungal metabolic unicity, either in opportunistic pathogens or commensals, may be exploited to develop novel therapeutic strategies. In doing so, we present our recent experience in different pathological settings that ultimately converge on relevant trans-kingdom metabolic differences.

2.
Sci Rep ; 13(1): 22692, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-38123809

RESUMO

Cystic fibrosis (CF) is an autosomal recessive disorder characterized by respiratory failure due to a vicious cycle of defective Cystic Fibrosis Transmembrane conductance Regulator (CFTR) function, chronic inflammation and recurrent bacterial and fungal infections. Although the recent introduction of CFTR correctors/potentiators has revolutionized the clinical management of CF patients, resurgence of inflammation and persistence of pathogens still posit a major concern and should be targeted contextually. On the background of a network-based selectivity that allows to target the same enzyme in the host and microbes with different outcomes, we focused on sphingosine-1-phosphate (S1P) lyase (SPL) of the sphingolipid metabolism as a potential candidate to uniquely induce anti-inflammatory and antifungal activities in CF. As a feasibility study, herein we show that interfering with S1P metabolism improved the immune response in a murine model of CF with aspergillosis while preventing germination of Aspergillus fumigatus conidia. In addition, in an early drug discovery process, we purified human and A. fumigatus SPL, characterized their biochemical and structural properties, and performed an in silico screening to identify potential dual species SPL inhibitors. We identified two hits behaving as competitive inhibitors of pathogen and host SPL, thus paving the way for hit-to-lead and translational studies for the development of drug candidates capable of restraining fungal growth and increasing antifungal resistance.


Assuntos
Fibrose Cística , Humanos , Animais , Camundongos , Fibrose Cística/tratamento farmacológico , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Estudos de Viabilidade , Inflamação , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico
3.
Biochim Biophys Acta Mol Cell Res ; 1870(8): 119554, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37524263

RESUMO

Hydroquinone, a potent toxic agent of cigarette smoke, damages retinal pigmented epithelial cells by triggering oxidative stress and mitochondrial dysfunction, two events causally related to the development and progression of retinal diseases. The inner mitochondrial membrane is enriched in cardiolipin, a phospholipid susceptible of oxidative modifications which determine cell-fate decision. Using ARPE-19 cell line as a model of retinal pigmented epithelium, we analyzed the potential involvement of cardiolipin in hydroquinone toxicity. Hydroquinone exposure caused an early concentration-dependent increase in mitochondrial reactive oxygen species, decrease in mitochondrial membrane potential, and rise in the rate of oxygen consumption not accompanied by changes in ATP levels. Despite mitochondrial impairment, cell viability was preserved. Hydroquinone induced cardiolipin translocation to the outer mitochondrial membrane, and an increase in the colocalization of the autophagosome adapter protein LC3 with mitochondria, indicating the induction of protective mitophagy. A prolonged hydroquinone treatment induced pyroptotic cell death by cardiolipin-mediated caspase-1 and gasdermin-D activation. Cardiolipin-specific antioxidants counteracted hydroquinone effects pointing out that cardiolipin can act as a mitochondrial "eat-me signal" or as a pyroptotic cell death trigger. Our results indicate that cardiolipin may act as a timer for the mitophagy to pyroptosis switch and propose cardiolipin-targeting compounds as promising approaches for the treatment of oxidative stress-related retinal diseases.


Assuntos
Cardiolipinas , Doenças Retinianas , Humanos , Cardiolipinas/metabolismo , Hidroquinonas/toxicidade , Hidroquinonas/metabolismo , Células Epiteliais/metabolismo , Doenças Retinianas/metabolismo
4.
EMBO Mol Med ; 15(4): e17033, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36647689

RESUMO

Gyrate atrophy of choroid and retina (GACR) is a chorioretinal degeneration caused by pathogenic variants in the gene encoding ornithine aminotransferase (OAT), an enzyme mainly expressed in liver. Affected patients have increased ornithine concentrations in blood and other body fluids and develop progressive constriction of vision fields leading to blindness. Current therapies are unsatisfactory and better treatments are highly needed. In two mouse models of OAT deficiency that recapitulates biochemical and retinal changes of GACR, we investigated the efficacy of an intravenously injected serotype 8 adeno-associated (AAV8) vector expressing OAT under the control of a hepatocyte-specific promoter. Following injections, OAT-deficient mice showed reductions of ornithine concentrations in blood and eye cups compared with control mice injected with a vector expressing green fluorescent protein. AAV-injected mice showed improved electroretinogram response and partial restoration of retinal structure up to one-year post-injection. In summary, hepatic OAT expression by AAV8 vector was effective at correction of hyperornithinemia and improved function and structure of the retina. In conclusion, this study provides proof-of-concept of efficacy of liver-directed AAV-mediated gene therapy of GACR.


Assuntos
Atrofia Girata , Degeneração Retiniana , Animais , Camundongos , Atrofia Girata/genética , Atrofia Girata/patologia , Ornitina-Oxo-Ácido Transaminase/genética , Ornitina-Oxo-Ácido Transaminase/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Ornitina/genética , Ornitina/metabolismo , Terapia Genética , Fígado/patologia
5.
Biochimie ; 202: 110-122, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35964771

RESUMO

AGXT1 encodes alanine:glyoxylate aminotransferase 1 (AGT1), a liver peroxisomal pyridoxal 5'-phosphate dependent-enzyme whose deficit causes Primary Hyperoxaluria Type 1 (PH1). PH1 is a rare disease characterized by overproduction of oxalate, first leading to kidney stones formation, and possibly evolving to life-threatening systemic oxalosis. A minority of PH1 patients is responsive to pyridoxine, while the option for non-responders is liver-kidney transplantation. Therefore, huge efforts are currently focused on the identification of new therapies, including the promising approaches based on RNA silencing recently approved. Many PH1-associated mutations are missense and lead to a variety of kinetic and/or folding defects on AGT1. In this context, the availability of a reliable in vitro disease model would be essential to better understand the phenotype of known or newly-identified pathogenic variants as well as to test novel drug candidates. Here, we took advantage of the CRISPR/Cas9 technology to specifically knock-out AGXT1 in HepG2 cells, a hepatoma-derived cell model exhibiting a conserved glyoxylate metabolism. AGXT1-KO HepG2 displayed null AGT1 expression and significantly reduced transaminase activity leading to an enhanced secretion of oxalate upon glycolate challenge. Known pathogenic AGT1 variants expressed in AGXT1-KO HepG2 cells showed alteration in both protein levels and specific transaminase activity, as well as a partial mitochondrial mistargeting when associated with a common polymorphism. Notably, pyridoxine treatment was able to partially rescue activity and localization of clinically-responsive variants. Overall, our data validate AGXT1-KO HepG2 cells as a novel cellular model to investigate PH1 pathophysiology, and as a platform for drug discovery and development.


Assuntos
Sistemas CRISPR-Cas , Piridoxina , Humanos , Células Hep G2 , Piridoxina/farmacologia , Transaminases/genética , Oxalatos , Fosfato de Piridoxal
6.
FASEB J ; 36(4): e22218, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35218567

RESUMO

An immunoregulatory role of stem cells, often mediated by their secretome, has been claimed by several studies. Stem cell-derived extracellular vesicles (EVs) are crucial components of the secretome. EVs, a heterogeneous group of membranous vesicles released by many cell types into the extracellular space, are now considered as an additional mechanism for intercellular communication. In this study, we aimed at investigating whether human amniotic stem cell-derived extracellular vesicles (HASC-EVs) were able to interfere with inflammasome activation in the THP-1 cell line. Two subsets of HASC-EVs were collected by sequential centrifugation, namely HASC-P10 and HASC-P100. We demonstrated that HASC-EVs were neither internalized into nor undertake a direct interaction with THP-1 cells. We showed that HASC-P10 and P100 were able to intrinsically produce ATP, which was further converted to adenosine by 5'-nucleotidase (CD73) and ectonucleoside triphosphate diphosphohydrolase-1 (CD39). We found that THP-1 cells conditioned with both types of HASC-EVs failed to activate the NLRP3/caspase-1/inflammasome platform in response to LPS and ATP treatment by a mechanism involving A2a adenosine receptor activation. These results support a role for HASC-EVs as independent metabolic units capable of modifying the cellular functions, leading to anti-inflammatory effects in monocytic cells.


Assuntos
Líquido Amniótico/citologia , Anti-Inflamatórios/farmacologia , Vesículas Extracelulares/metabolismo , Inflamassomos/antagonistas & inibidores , Inflamação/prevenção & controle , Monócitos/citologia , Células-Tronco/citologia , Adenosina/metabolismo , Líquido Amniótico/metabolismo , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Monócitos/metabolismo , Antagonistas de Receptores Purinérgicos P1/farmacologia , Receptores Purinérgicos P1/química , Receptores Purinérgicos P1/metabolismo , Células-Tronco/metabolismo , Células THP-1
7.
J Clin Invest ; 132(2)2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34847078

RESUMO

Autophagy selectively degrades aggregation-prone misfolded proteins caused by defective cellular proteostasis. However, the complexity of autophagy may prevent the full appreciation of how its modulation could be used as a therapeutic strategy in disease management. Here, we define a molecular pathway through which recombinant IL-1 receptor antagonist (IL-1Ra, anakinra) affects cellular proteostasis independently from the IL-1 receptor (IL-1R1). Anakinra promoted H2O2-driven autophagy through a xenobiotic sensing pathway involving the aryl hydrocarbon receptor that, activated through the indoleamine 2,3-dioxygenase 1-kynurenine pathway, transcriptionally activated NADPH oxidase 4 independent of the IL-1R1. By coupling the mitochondrial redox balance to autophagy, anakinra improved the dysregulated proteostasis network in murine and human cystic fibrosis. We anticipate that anakinra may represent a therapeutic option in addition to its IL-1R1-dependent antiinflammatory properties by acting at the intersection of mitochondrial oxidative stress and autophagy with the capacity to restore conditions in which defective proteostasis leads to human disease.


Assuntos
Autofagia/efeitos dos fármacos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteostase/efeitos dos fármacos , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , Oxirredução/efeitos dos fármacos
8.
Eur J Med Chem ; 206: 112717, 2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-32823008

RESUMO

Cystic fibrosis (CF) is a rare genetic disorder caused by a defect in the ion channel Cystic Fibrosis Transmembrane conductance Regulator (CFTR), resulting in ionic imbalance of surface fluid. Although affecting multiple organs, the progressive deterioration of respiratory function by recurrent infections and chronic inflammation represents the main cause of morbidity and mortality in CF patients. The development of modulators targeting the basic defect of CFTR has represented a major breakthrough in CF therapy, but the impact on inflammation has remained enigmatic. The emerging scenario taking hold in the field points to inflammation as a major, somehow missed, therapeutic target for prevention of lung decline. Not surprisingly, the development of anti-inflammatory drugs is taking its share in the drug development pipeline. But the path is not straightforward and targeting inflammation should be balanced with the increased risk of infection. The strategy to restore the homeostatic regulation of inflammation to efficiently respond to infection while preventing lung damage needs to be based on identifying and targeting endogenous immunoregulatory pathways that are defective in CF. We herein provide an overview of anti-inflammatory drugs currently approved or under investigation in CF patients, and present our recent studies on how the knowledge on defective immune pathways in CF may translate into innovative and selective anti-inflammatory therapeutics. Through the discovery of naturally occurring molecules or their synthetic mimics, this review emphasizes the critical importance of selectively targeting key inflammatory pathways to preserve immunocompetence in CF patients.


Assuntos
Fibrose Cística/tratamento farmacológico , Terapia de Alvo Molecular/métodos , Animais , Fibrose Cística/complicações , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Descoberta de Drogas , Humanos , Inflamação/complicações , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia
9.
Hum Mol Genet ; 28(1): 1-15, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30215702

RESUMO

Most pathogenic missense mutations cause specific molecular phenotypes through protein destabilization. However, how protein destabilization is manifested as a given molecular phenotype is not well understood. We develop here a structural and energetic approach to describe mutational effects on specific traits such as function, regulation, stability, subcellular targeting or aggregation propensity. This approach is tested using large-scale experimental and structural perturbation analyses in over thirty mutations in three different proteins (cancer-associated NQO1, transthyretin related with amyloidosis and AGT linked to primary hyperoxaluria type I) and comprising five very common pathogenic mechanisms (loss-of-function and gain-of-toxic function aggregation, enzyme inactivation, protein mistargeting and accelerated degradation). Our results revealed that the magnitude of destabilizing effects and, particularly, their propagation through the structure to promote disease-associated conformational states largely determine the severity and molecular mechanisms of disease-associated missense mutations. Modulation of the structural perturbation at a mutated site is also shown to cause switches between different molecular phenotypes. When very common disease-associated missense mutations were investigated, we also found that they were not among the most deleterious possible missense mutations at those sites, and required additional contributions from codon bias and effects of CpG sites to explain their high frequency in patients. Our work sheds light on the molecular basis of pathogenic mechanisms and genotype-phenotype relationships, with implications for discriminating between pathogenic and neutral changes within human genome variability from whole genome sequencing studies.


Assuntos
Mutação de Sentido Incorreto/fisiologia , Proteínas/genética , Relação Estrutura-Atividade , Animais , Biologia Computacional/métodos , Biologia Computacional/estatística & dados numéricos , Doença , Humanos , Mutação , Mutação de Sentido Incorreto/genética , Patologia , Fenótipo , Conformação Proteica , Proteínas/fisiologia
10.
Curr Protein Pept Sci ; 19(8): 805-812, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29708066

RESUMO

Protein function is dependent on assumption of the correct three-dimensional structure, achieved through the folding process. As a central element in ensuring cellular homeostasis, proteostasis i.e. the control of correct protein folding, trafficking and degradation, is a highly regulated process ensured by three integrated molecular pathways: i) the unfolded protein response (UPR) which is activated by the engulfment of misfolded proteins and results in protein re-folding through the expression of chaperones; ii) the ubiquitin-proteasome system (UPS) which 'flags' misfolded proteins with ubiquitin, directing them to the 26S proteasome for proteolytic degradation; iii) autophagy that, through lysosomes, removes misfolded or aggregated proteins. All three of these proteostatic controls can be impaired by the aging process and by pathological mutations highlighting the potential role of proteostasis in conditions associated with aging such as neurodegeneration, type 2 diabetes and cancer. Indeed, neurodegenerative diseases are characterised by an interconnected triumvirate of deregulated proteostasis, neuroinflammation (i.e. the uncontrolled activation of microglial cells), and oxidative stress (i.e. the unbuffered increase in reactive oxygen species). The transcription factor Nrf2, classically associated with protection against oxidative stress, can also modulate the UPR, UPS and autophagy, while inhibiting the activation of NF-kB, the key transcription factor of the inflammatory response. In this review we focus on recent data from our laboratory and others demonstrating that the protective Nrf2 pathway can be activated by the endogenous cyclic dipeptide (His-Pro), thereby driving neuroprotective effects in different pathological settings. In this context we discuss the possible utility of clyclo (His-Pro) as a promising future therapeutic option for protein misfolding disorders.


Assuntos
Doenças Neurodegenerativas/metabolismo , Peptídeos Cíclicos/metabolismo , Piperazinas/metabolismo , Proteostase , Animais , Autofagia , Morte Celular , Sobrevivência Celular , Humanos , NF-kappa B/metabolismo , Doenças Neurodegenerativas/terapia , Estresse Oxidativo , Conformação Proteica , Dobramento de Proteína , Proteólise , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/terapia , Transdução de Sinais
11.
Free Radic Biol Med ; 117: 191-201, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29427792

RESUMO

STAT1 and STAT3 are two transcription factors involved in a lot of cellular functions such as immune response, proliferation, apoptosis, and cell survival. A number of literature evidences described a yin-yang relationship between activation of STAT1 and STAT3 in neurodegenerative disorders where STAT1 exerts a pro-apoptotic effect whereas STAT3 shows neuroprotective properties through the inhibition of apoptosis. Although the role of oxidative-stress in the pathogenesis of neurodegeneration is clearly described, its influence in the regulation of these pathways is poorly understood. Herein, we demonstrate that H2O2 rapidly induces phosphorylation of STAT1 whereas it is not able to influence phosphorylation of STAT3 in mouse microglia BV2 cells. The analysis of the molecular mechanism of STATs signaling reveals that H2O2 induces S-glutathionylation of both STAT1 and STAT3. The same post-translational event exerts an opposing role in the regulation of STAT1 and STAT3 signaling. These data not only confirm redox sensibility of STAT3 signaling but also reveal for the first time that STAT1 is susceptible to redox regulation. A deep study of the molecular mechanism of STAT1 redox regulation, identifies Cys324 and Cys492 as the main targets of S-glutathionylation and confirms that S-glutathionylation does not impair JAK2 mediated STAT1 tyrosine phosphorylation. These results demonstrate that both phosphorylation and glutathionylation contribute to activation of STAT1 during oxidative stress and underline that the same post-translation event exerts an opposing role in the regulation of STAT1 and STAT3 signaling in microglia cells.


Assuntos
Ativação Enzimática/fisiologia , Microglia/metabolismo , Estresse Oxidativo/fisiologia , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Linhagem Celular , Glutationa/metabolismo , Peróxido de Hidrogênio/farmacologia , Camundongos , Oxidantes/farmacologia , Fosforilação , Transdução de Sinais/fisiologia
12.
Sci Rep ; 7(1): 11704, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28916765

RESUMO

The alanine:glyoxylate aminotransferase (AGT), a hepatocyte-specific pyridoxal-5'-phosphate (PLP) dependent enzyme, transaminates L-alanine and glyoxylate to glycine and pyruvate, thus detoxifying glyoxylate and preventing pathological oxalate precipitation in tissues. In the widely accepted catalytic mechanism of the aminotransferase family, the lysine binding to PLP acts as a catalyst in the stepwise 1,3-proton transfer, interconverting the external aldimine to ketimine. This step requires protonation by a conserved aspartate of the pyridine nitrogen of PLP to enhance its ability to stabilize the carbanionic intermediate. The aspartate residue is also responsible for a significant geometrical distortion of the internal aldimine, crucial for catalysis. We present the structure of human AGT in which complete X-ray photoreduction of the Schiff base has occurred. This result, together with two crystal structures of the conserved aspartate pathogenic variant (D183N) and the molecular modeling of the transaldimination step, led us to propose that an interplay of opposite forces, which we named spring mechanism, finely tunes PLP geometry during catalysis and is essential to move the external aldimine in the correct position in order for the 1,3-proton transfer to occur.


Assuntos
Domínio Catalítico/efeitos da radiação , Transaminases/metabolismo , Alanina , Catálise , Humanos , Lisina/metabolismo , Modelos Moleculares , Estrutura Molecular , Prótons , Fosfato de Piridoxal/metabolismo , Bases de Schiff/química , Bases de Schiff/efeitos da radiação , Raios X
13.
Cell Rep ; 20(7): 1667-1680, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28813677

RESUMO

The interleukin 17 (IL-17) cytokine and receptor family is central to antimicrobial resistance and inflammation in the lung. Mice lacking IL-17A, IL-17F, or the IL-17RA subunit were compared with wild-type mice for susceptibility to airway inflammation in models of infection and allergy. Signaling through IL-17RA was required for efficient microbial clearance and prevention of allergy; in the absence of IL-17RA, signaling through IL-17RC on epithelial cells, predominantly by IL-17F, significantly exacerbated lower airway Aspergillus or Pseudomonas infection and allergic airway inflammation. In contrast, following infection with the upper respiratory pathogen Staphylococcus aureus, the IL-17F/IL-17RC axis mediated protection. Thus, IL-17A and IL-17F exert distinct biological effects during pulmonary infection; the IL-17F/IL-17RC signaling axis has the potential to significantly worsen pathogen-associated inflammation of the lower respiratory tract in particular, and should be investigated further as a therapeutic target for treating pathological inflammation in the lung.


Assuntos
Aspergilose/imunologia , Hipersensibilidade/imunologia , Interleucina-17/imunologia , Infecções por Pseudomonas/imunologia , Receptores de Interleucina-17/imunologia , Infecções Estafilocócicas/imunologia , Animais , Aspergilose/genética , Aspergilose/microbiologia , Aspergilose/patologia , Aspergillus/imunologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Hipersensibilidade/genética , Hipersensibilidade/microbiologia , Hipersensibilidade/patologia , Interleucina-17/deficiência , Interleucina-17/genética , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Pseudomonas/imunologia , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Receptores de Interleucina-17/deficiência , Receptores de Interleucina-17/genética , Mucosa Respiratória/imunologia , Mucosa Respiratória/microbiologia , Mucosa Respiratória/patologia , Transdução de Sinais , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/imunologia
14.
Nanomedicine ; 13(3): 897-907, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27993722

RESUMO

Alanine:glyoxylate aminotransferase (AGT) is a liver peroxisomal enzyme whose deficit causes the rare disorder Primary Hyperoxaluria Type I (PH1). We now describe the conjugation of poly(ethylene glycol)-co-poly(L-glutamic acid) (PEG-PGA) block-co-polymer to AGT via the formation of disulfide bonds between the polymer and solvent-exposed cysteine residues of the enzyme. PEG-PGA conjugation did not affect AGT structural/functional properties and allowed the enzyme to be internalized in a cellular model of PH1 and to restore glyoxylate-detoxification. The insertion of the C387S/K390S amino acid substitutions, known to favor interaction with the peroxisomal import machinery, reduced conjugation efficiency, but endowed conjugates with the ability to reach the peroxisomal compartment. These results, along with the finding that conjugates are hemocompatible, stable in plasma, and non-immunogenic, hold promise for the development of polypeptide-based AGT conjugates as a therapeutic option for PH1 patients and represent the base for applications to other diseases related to deficits in peroxisomal proteins.


Assuntos
Sistemas de Liberação de Medicamentos , Hiperoxalúria Primária/tratamento farmacológico , Peroxissomos/metabolismo , Polietilenoglicóis/química , Ácido Poliglutâmico/análogos & derivados , Transaminases/administração & dosagem , Transaminases/química , Substituição de Aminoácidos , Animais , Células CHO , Cricetulus , Terapia Enzimática , Glioxilatos/metabolismo , Humanos , Hiperoxalúria Primária/enzimologia , Hiperoxalúria Primária/metabolismo , Modelos Moleculares , Peroxissomos/efeitos dos fármacos , Transaminases/genética , Transaminases/farmacocinética
15.
Sci Rep ; 6: 35277, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27752063

RESUMO

Low plasma concentrations of L-homoarginine are associated with an increased risk of cardiovascular events, while homoarginine supplementation is protective in animal models of metabolic syndrome and stroke. Catabolism of homoarginine is still poorly understood. Based on the recent findings from a Genome Wide Association Study we hypothesized that homoarginine can be metabolized by alanine:glyoxylate aminotransferase 2 (AGXT2). We purified human AGXT2 from tissues of AGXT2 transgenic mice and demonstrated its ability to metabolize homoarginine to 6-guanidino-2-oxocaproic acid (GOCA). After incubation of HepG2 cells overexpressing AGXT2 with isotope-labeled homoarginine-d4 we were able to detect labeled GOCA in the medium. We injected wild type mice with labeled homoarginine and detected labeled GOCA in the plasma. We found that AGXT2 knockout (KO) mice have higher homoarginine and lower GOCA plasma levels as compared to wild type mice, while the reverse was true for AGXT2 transgenic (Tg) mice. In summary, we experimentally proved the presence of a new pathway of homoarginine catabolism - its transamination by AGXT2 with formation of GOCA and demonstrated that endogenous AGXT2 is required for maintenance of homoarginine levels in mice. Our findings may lead to development of novel therapeutic approaches for cardiovascular pathologies associated with homoarginine deficiency.


Assuntos
Doenças Cardiovasculares/sangue , Homoarginina/sangue , Síndrome Metabólica/genética , Acidente Vascular Cerebral/sangue , Transaminases/genética , Animais , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/patologia , Estudo de Associação Genômica Ampla , Células Hep G2 , Homoarginina/genética , Humanos , Redes e Vias Metabólicas/genética , Síndrome Metabólica/sangue , Síndrome Metabólica/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fatores de Risco , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia
16.
Mol Oncol ; 10(7): 1008-29, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27118659

RESUMO

Mutations in TP53 gene play a pivotal role in tumorigenesis and cancer development. Here, we report that gain-of-function mutant p53 proteins inhibit the autophagic pathway favoring antiapoptotic effects as well as proliferation of pancreas and breast cancer cells. We found that mutant p53 significantly counteracts the formation of autophagic vesicles and their fusion with lysosomes throughout the repression of some key autophagy-related proteins and enzymes as BECN1 (and P-BECN1), DRAM1, ATG12, SESN1/2 and P-AMPK with the concomitant stimulation of mTOR signaling. As a paradigm of this mechanism, we show that atg12 gene repression was mediated by the recruitment of the p50 NF-κB/mutant p53 protein complex onto the atg12 promoter. Either mutant p53 or p50 NF-κB depletion downregulates atg12 gene expression. We further correlated the low expression levels of autophagic genes (atg12, becn1, sesn1, and dram1) with a reduced relapse free survival (RFS) and distant metastasis free survival (DMFS) of breast cancer patients carrying TP53 gene mutations conferring a prognostic value to this mutant p53-and autophagy-related signature. Interestingly, the mutant p53-driven mTOR stimulation sensitized cancer cells to the treatment with the mTOR inhibitor everolimus. All these results reveal a novel mechanism through which mutant p53 proteins promote cancer cell proliferation with the concomitant inhibition of autophagy.


Assuntos
Autofagia/efeitos dos fármacos , Proteínas Mutantes/metabolismo , Neoplasias/patologia , Inibidores de Proteínas Quinases/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Adenilato Quinase/metabolismo , Apoptose/efeitos dos fármacos , Proteína 12 Relacionada à Autofagia/metabolismo , Sequência de Bases , Proteína Beclina-1/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Everolimo/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Modelos Biológicos , Neoplasias/genética , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Fator de Transcrição RelA/metabolismo
17.
ACS Chem Biol ; 9(8): 1885-93, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24941337

RESUMO

STAT3 is a latent transcription factor that promotes cell survival and proliferation and is often constitutively active in cancers. Although many reports provide evidence that STAT3 is a direct target of oxidative stress, its redox regulation is poorly understood. Under oxidative conditions STAT3 activity can be modulated by S-glutathionylation, a reversible redox modification of cysteine residues. This suggests the possible cross-talk between phosphorylation and glutathionylation and points out that STAT3 is susceptible to redox regulation. Recently, we reported that decreasing the GSH content in different cell lines induces inhibition of STAT3 activity through the reversible oxidation of thiol groups. In the present work, we demonstrate that GSH/diamide treatment induces S-glutathionylation of STAT3 in the recombinant purified form. This effect was completely reversed by treatment with the reducing agent dithiothreitol, indicating that S-glutathionylation of STAT3 was related to formation of protein-mixed disulfides. Moreover, addition of the bulky negatively charged GSH moiety impairs JAK2-mediated STAT3 phosphorylation, very likely interfering with tyrosine accessibility and thus affecting protein structure and function. Mass mapping analysis identifies two glutathionylated cysteine residues, Cys328 and Cys542, within the DNA-binding domain and the linker domain, respectively. Site direct mutagenesis and in vitro kinase assay confirm the importance of both cysteine residues in the complex redox regulatory mechanism of STAT3. Cells expressing mutant were resistant in this regard. The data presented herein confirmed the occurrence of a redox-dependent regulation of STAT3, identified the more redox-sensitive cysteines within STAT3 structure, and may have important implications for development of new drugs.


Assuntos
Cisteína/metabolismo , Glutationa/metabolismo , Fator de Transcrição STAT3/metabolismo , Células HeLa , Humanos , Oxirredução , Fosforilação , Fator de Transcrição STAT3/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
18.
ChemMedChem ; 9(7): 1501-11, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24616267

RESUMO

Cystalysin from Treponema denticola is a pyridoxal 5'-phosphate dependent lyase that catalyzes the formation of pyruvate, ammonia, and sulfide from cysteine. It is a virulence factor in adult periodontitis because its reaction contributes to hemolysis, which sustains the pathogen. Therefore, it was proposed as a potential antimicrobial target. To identify specific inhibitors by structure-based in silico methods, we first validated the crystal structure of cystalysin as a reliable starting point for the design of ligands. By using single-crystal absorption microspectrophotometry, we found that the enzyme in the crystalline state, with respect to that in solution, exhibits: 1) the same absorption spectra for the catalytic intermediates, 2) a close pKa value for the residue controlling the keto enamine ionization, and 3) similar reactivity with glycine, L-serine, L-methionine, and the nonspecific irreversible inhibitor aminoethoxyvinylglycine. Next, we screened in silico a library of 9357 compounds with the Fingerprints for Ligands and Proteins (FLAP) software, by using the three-dimensional structure of cystalysin as a template. From the library, 17 compounds were selected and experimentally evaluated by enzyme assays and spectroscopic methods. Two compounds were found to competitively inhibit recombinant T. denticola cystalysin, with inhibition constant (Ki ) values of 25 and 37 µM. One of them exhibited a minimum inhibitory concentration (MIC) value of 64 µg mL(-1) on Moraxella catarrhalis ATCC 23246, which proves its ability to cross bacterial membranes.


Assuntos
Cistationina gama-Liase/antagonistas & inibidores , Inibidores Enzimáticos/química , Treponema denticola/enzimologia , Antibacterianos/química , Antibacterianos/farmacologia , Sítios de Ligação , Domínio Catalítico , Cistationina gama-Liase/genética , Cistationina gama-Liase/metabolismo , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Periodontite/tratamento farmacológico , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/uso terapêutico , Treponema denticola/efeitos dos fármacos
19.
Clin Biochem ; 47(3): 158-65, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24355692

RESUMO

The biologically active form of the B6 vitamers is pyridoxal 5'-phosphate (PLP), which plays a coenzymatic role in several distinct enzymatic activities ranging from the synthesis, interconversion and degradation of amino acids to the replenishment of one-carbon units, synthesis and degradation of biogenic amines, synthesis of tetrapyrrolic compounds and metabolism of amino-sugars. In the catalytic process of PLP-dependent enzymes, the substrate amino acid forms a Schiff base with PLP and the electrophilicity of the PLP pyridine ring plays important roles in the subsequent catalytic steps. While the essential role of PLP in the acquisition of biological activity of many proteins is long recognized, the finding that some PLP-enzymes require the coenzyme for refolding in vitro points to an additional role of PLP as a chaperone in the folding process. Mutations in the genes encoding PLP-enzymes are causative of several rare inherited diseases. Patients affected by some of these diseases (AADC deficiency, cystathionuria, homocystinuria, gyrate atrophy, primary hyperoxaluria type 1, xanthurenic aciduria, X-linked sideroblastic anaemia) can benefit, although at different degrees, from the administration of pyridoxine, a PLP precursor. The effect of the coenzyme is not limited to mutations that affect the enzyme-coenzyme interaction, but also to those that cause folding defects, reinforcing the idea that PLP could play a chaperone role and improve the folding efficiency of misfolded variants. In this review, recent biochemical and cell biology studies highlighting the chaperoning activity of the coenzyme on folding-defective variants of PLP-enzymes associated with rare diseases are presented and discussed.


Assuntos
Coenzimas/metabolismo , Erros Inatos do Metabolismo/enzimologia , Chaperonas Moleculares/metabolismo , Fosfato de Piridoxal/metabolismo , Vitamina B 6/metabolismo , Animais , Coenzimas/genética , Humanos , Erros Inatos do Metabolismo/genética , Chaperonas Moleculares/genética , Dobramento de Proteína , Deficiências na Proteostase/enzimologia , Deficiências na Proteostase/genética , Fosfato de Piridoxal/genética , Vitamina B 6/genética
20.
Biomed Res Int ; 2013: 701536, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24106714

RESUMO

The emergence of antibiotic resistance in microbial pathogens requires the identification of new antibacterial drugs. The biosynthesis of methionine is an attractive target because of its central importance in cellular metabolism. Moreover, most of the steps in methionine biosynthesis pathway are absent in mammals, lowering the probability of unwanted side effects. Herein, detailed biochemical characterization of one enzyme required for methionine biosynthesis, a pyridoxal-5'-phosphate (PLP-) dependent C-S lyase from Corynebacterium diphtheriae, a pathogenic bacterium that causes diphtheria, has been performed. We overexpressed the protein in E. coli and analyzed substrate specificity, pH dependence of steady state kinetic parameters, and ligand-induced spectral transitions of the protein. Structural comparison of the enzyme with cystalysin from Treponema denticola indicates a similarity in overall folding. We used site-directed mutagenesis to highlight the importance of active site residues Tyr55, Tyr114, and Arg351, analyzing the effects of amino acid replacement on catalytic properties of enzyme. Better understanding of the active site of C. diphtheriae C-S lyase and the determinants of substrate and reaction specificity from this work will facilitate the design of novel inhibitors as antibacterial therapeutics.


Assuntos
Corynebacterium diphtheriae/enzimologia , Difteria/tratamento farmacológico , Farmacorresistência Bacteriana/genética , Liases/química , Catálise , Domínio Catalítico , Corynebacterium diphtheriae/genética , Cistationina gama-Liase/química , Difteria/enzimologia , Difteria/patologia , Escherichia coli , Regulação Enzimológica da Expressão Gênica , Humanos , Cinética , Liases/genética , Metionina/biossíntese , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA