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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(11)2023 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-37298727

RESUMO

Gut dysbiosis has been involved in the pathogenesis and progression of Parkinson's disease (PD), but the mechanisms through which gut microbiota (GM) exerts its influences deserve further study. Recently, we proposed a two-hit mouse model of PD in which ceftriaxone (CFX)-induced dysbiosis amplifies the neurodegenerative phenotype generated by striatal 6-hydroxydopamine (6-OHDA) injection in mice. Low GM diversity and the depletion of key gut colonizers and butyrate producers were the main signatures of GM alteration in this model. Here, we used the phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt2) to unravel candidate pathways of cell-to-cell communication associated with dual-hit mice and potentially involved in PD progression. We focused our analysis on short-chain fatty acids (SCFAs) metabolism and quorum sensing (QS) signaling. Based on linear discriminant analysis, combined with the effect size results, we found increased functions linked to pyruvate utilization and a depletion of acetate and butyrate production in 6-OHDA+CFX mice. The specific arrangement of QS signaling as a possible result of the disrupted GM structure was also observed. With this exploratory study, we suggested a scenario in which SCFAs metabolism and QS signaling might represent the effectors of gut dysbiosis potentially involved in the designation of the functional outcomes that contribute to the exacerbation of the neurodegenerative phenotype in the dual-hit animal model of PD.


Assuntos
Doença de Parkinson , Camundongos , Animais , Doença de Parkinson/metabolismo , Disbiose/metabolismo , Filogenia , Oxidopamina , Butiratos
2.
Int J Mol Sci ; 23(12)2022 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-35742813

RESUMO

Recent evidence highlights Parkinson's disease (PD) initiation in the gut as the prodromal phase of neurodegeneration. Gut impairment due to microbial dysbiosis could affect PD pathogenesis and progression. Here, we propose a two-hit model of PD through ceftriaxone (CFX)-induced dysbiosis and gut inflammation before the 6-hydroxydopamine (6-OHDA) intrastriatal injection to mimic dysfunctional gut-associated mechanisms preceding PD onset. Therefore, we showed that dysbiosis and gut damage amplified PD progression, worsening motor deficits induced by 6-OHDA up to 14 days post intrastriatal injection. This effect was accompanied by a significant increase in neuronal dopaminergic loss (reduced tyrosine hydroxylase expression and increased Bcl-2/Bax ratio). Notably, CFX pretreatment also enhanced systemic and colon inflammation of dual-hit subjected mice. The exacerbated inflammatory response ran in tandem with a worsening of colonic architecture and gut microbiota perturbation. Finally, we demonstrated the beneficial effect of post-biotic sodium butyrate in limiting at once motor deficits, neuroinflammation, and colon damage and re-shaping microbiota composition in this novel dual-hit model of PD. Taken together, the bidirectional communication of the microbiota-gut-brain axis and the recapitulation of PD prodromal/pathogenic features make this new paradigm a useful tool for testing or repurposing new multi-target compounds in the treatment of PD.


Assuntos
Disbiose , Doença de Parkinson , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Butiratos/farmacologia , Butiratos/uso terapêutico , Disbiose/patologia , Inflamação/patologia , Camundongos , Oxidopamina , Doença de Parkinson/metabolismo
3.
Epilepsia ; 62(2): 529-541, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33428780

RESUMO

OBJECTIVE: A large number of studies have highlighted the important role of the gut microbiota in the pathophysiology of neurological disorders, suggesting that its manipulation might serve as a treatment strategy. We hypothesized that the gut microbiota participates in absence seizure development and maintenance in the WAG/Rij rat model and tested this hypothesis by evaluating potential gut microbiota and intestinal alterations in the model, as well as measuring the impact of microbiota manipulation using fecal microbiota transplantation (FMT). METHODS: Initially, gut microbiota composition and intestinal histology of WAG/Rij rats (a well-recognized genetic model of absence epilepsy) were studied at 1, 4, and 8 months of age in comparison to nonepileptic Wistar rats. Subsequently, in a second set of experiments, at 6 months of age, untreated Wistar or WAG/Rij rats treated with ethosuximide (ETH) were used as gut microbiota donors for FMT in WAG/Rij rats, and electroencephalographic (EEG) recordings were obtained over 4 weeks. At the end of FMT, stool and gut samples were collected, absence seizures were measured on EEG recordings, and microbiota analysis and histopathological examinations were performed. RESULTS: Gut microbiota analysis showed differences in beta diversity and specific phylotypes at all ages considered and significant variances in the Bacteroidetes/Firmicutes ratio between Wistar and WAG/Rij rats. FMT, from both Wistar and ETH-treated WAG/Rij donors to WAG/Rij rats, significantly decreased the number and duration of seizures. Histological results indicated that WAG/Rij rats were characterized by intestinal villi disruption and inflammatory infiltrates already at 1 month of age, before seizure occurrence; FMT partially restored intestinal morphology while also significantly modifying gut microbiota and concomitantly reducing absence seizures. SIGNIFICANCE: Our results demonstrate for the first time that the gut microbiota is modified and contributes to seizure occurrence in a genetic animal model of absence epilepsy and that its manipulation may be a suitable therapeutic target for absence seizure management.


Assuntos
Antibacterianos/farmacologia , Anticonvulsivantes/farmacologia , Epilepsia Tipo Ausência/microbiologia , Transplante de Microbiota Fecal , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Animais , Bacteroidetes , Butiratos/metabolismo , Colo/patologia , DNA Bacteriano/análise , DNA Ribossômico/genética , Modelos Animais de Doenças , Eletroencefalografia , Epilepsia Tipo Ausência/genética , Epilepsia Tipo Ausência/fisiopatologia , Epilepsia Tipo Ausência/terapia , Etossuximida/farmacologia , Ácidos Graxos Voláteis/metabolismo , Firmicutes , Motilidade Gastrointestinal , Haptoglobinas/metabolismo , Íleo/patologia , Propionatos/metabolismo , Precursores de Proteínas/metabolismo , Proteobactérias , Ratos , Ratos Wistar , Convulsões/genética , Convulsões/microbiologia , Convulsões/fisiopatologia
4.
Pflugers Arch ; 472(7): 899-909, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32577860

RESUMO

Investigating the Shaker-related K+ channel Kv1.1, the dysfunction of which is responsible for episodic ataxia 1 (EA1), at the functional and molecular level provides valuable understandings on normal channel dynamics, structural correlates underlying voltage-gating, and disease-causing mechanisms. Most studies focused on apparently functional amino acid residues composing voltage-gated K+ channels, neglecting the simplest ones. Glycine at position 311 of Kv1.1 is highly conserved both evolutionarily and within the Kv channel superfamily, is located in a region functionally relevant (the S4-S5 linker), and results in overt disease when mutated (p.G311D). By mutating the G311 residue to aspartate, we show here that the channel voltage-gating, activation, deactivation, inactivation, and window currents are markedly affected. In silico, modeling shows this glycine residue is strategically placed at one end of the linker helix which must be free to both bend and move past other portions of the protein during the channel's opening and closing. This is befitting of a glycine residue as its small neutral side chain allows for movement unhindered by interaction with any other amino acid. Results presented reveal the crucial importance of a distinct glycine residue, within the S4-S5 linker, in the voltage-dependent electromechanical coupling that control channel gating.


Assuntos
Aminoácidos/metabolismo , Ativação do Canal Iônico/fisiologia , Canal de Potássio Kv1.1/genética , Sequência de Aminoácidos , Animais , Ataxia/metabolismo , Ataxia/patologia , Xenopus laevis/metabolismo
5.
Brain Behav Immun ; 74: 166-175, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30193877

RESUMO

Autism spectrum disorders (ASD) are a group of heterogeneous neurodevelopmental conditions characterized by impaired social interaction, and repetitive stereotyped behaviours. Interestingly, functional and inflammatory gastrointestinal diseases are often reported as a comorbidity in ASDs, indicating gut-brain axis as a novel emerging approach. Recently, a central role for peroxisome-proliferator activated receptor (PPAR)-α has been addressed in neurological functions, associated with the behaviour. Among endogenous lipids, palmitoylethanolamide (PEA), a PPAR-α agonist, has been extensively studied for its anti-inflammatory effects both at central and peripheral level. Based on this background, the aim of this study was to investigate the pharmacological effects of PEA on autistic-like behaviour of BTBR T+tf/J mice and to shed light on the contributing mechanisms. Our results showed that PEA reverted the altered behavioural phenotype of BTBR mice, and this effect was contingent to PPAR-α activation. Moreover, PEA was able to restore hippocampal BDNF signalling pathway, and improve mitochondrial dysfunction, both pathological aspects, known to be consistently associated with ASDs. Furthermore, PEA reduced the overall inflammatory state of BTBR mice, reducing the expression of pro-inflammatory cytokines at hippocampal, serum, and colonic level. The analysis of gut permeability and the expression of colonic tight junctions showed a reduction of leaky gut in PEA-treated BTBR mice. This finding together with PEA effect on gut microbiota composition suggests an involvement of microbiota-gut-brain axis. In conclusion, our results demonstrated a therapeutic potential of PEA in limiting ASD symptoms, through its pleiotropic mechanism of action, supporting neuroprotection, anti-inflammatory effects, and the modulation of gut-brain axis.


Assuntos
Transtorno do Espectro Autista/tratamento farmacológico , Transtorno do Espectro Autista/metabolismo , Etanolaminas/farmacologia , Ácidos Palmíticos/farmacologia , Amidas , Animais , Transtorno Autístico/tratamento farmacológico , Transtorno Autístico/metabolismo , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Colo/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Etanolaminas/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , PPAR alfa/efeitos dos fármacos , PPAR alfa/metabolismo , Ácidos Palmíticos/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Med Mycol ; 56(8): 987-993, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29462476

RESUMO

Cytochrome P450 CYP1A1 and CYP1B1 enzymes are regulated by the aryl hydrocarbon receptor (AhR), a transcription factor activated by a variety of ligands among which Malassezia metabolites. In this study, we analyzed the modulation of CYP1A1, CYP1B1, and AhR in human keratinocytes infected with different strains of Malassezia pachydermatis, as well as the upregulation of some genes involved in the epidermal homeostasis. We demonstrated that all the strains induced AhR activation and its nuclear translocation in HaCaT cells infected for 24 h, compared to untreated cells. The expression of CYP1A1 and CYP1B1, prototypical markers of the AhR signaling pathway, were upregulated with the level of CYP1A1 mRNA approximately 100-fold greater than that for CYP1B1. Filaggrin, involucrin, and TGaseI, proteins involved in epidermal differentiation, were all modulated by Malassezia pachydermatis strains, with the strongest induction observed for filaggrin. By contrast, quinone oxidoreductase 1 (NQO1), which is part of the antioxidant defense system involved in detoxification, was not modulated in our experimental model. In conclusions, our findings suggest that Malassezia pachydermatis infection of human keratinocytes induces activation of the AhR, and increases the expression of its responsive genes and markers of epidermal differentiation, paving the way for occurrence/exacerbation of pathological skin conditions.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Citocromo P-450 CYP1A1/biossíntese , Queratinócitos/metabolismo , Queratinócitos/microbiologia , Malassezia/crescimento & desenvolvimento , Receptores de Hidrocarboneto Arílico/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1B1/biossíntese , Citocromo P-450 CYP1B1/genética , Proteínas Filagrinas , Perfilação da Expressão Gênica , Humanos , RNA Mensageiro/análise , RNA Mensageiro/genética , Receptores de Hidrocarboneto Arílico/genética , Transcrição Gênica
7.
Appl Environ Microbiol ; 83(19)2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28733284

RESUMO

We recently demonstrated that cow's milk fermented with the probiotic Lactobacillus paracasei CBA L74 (FM-CBAL74) reduces the incidence of respiratory and gastrointestinal tract infections in young children attending school. This effect apparently derives from a complex regulation of non-immune and immune protective mechanisms. We investigated whether FM-CBAL74 could regulate gut microbiota composition and butyrate production. We randomly selected 20 healthy children (12 to 48 months) from the previous randomized controlled trial, before (t0) and after 3 months (t3) of dietary treatment with FM-CBAL74 (FM) or placebo (PL). Fecal microbiota was profiled using 16S rRNA gene amplicon sequencing, and the fecal butyrate concentration was also measured. Microbial alpha and beta diversities were not significantly different between groups prior to treatment. FM-CBAL74 but not PL treatment increased the relative abundance of Lactobacillus Individual Blautia, Roseburia, and Faecalibacterium oligotypes were associated with FM-CBAL74 treatment and demonstrated correlative associations with immune biomarkers. Accordingly, PICRUSt analysis predicted an increase in the proportion of genes involved in butyrate production pathways, consistent with an increase in fecal butyrate observed only in the FM group. Dietary supplementation with FM-CBAL74 induces specific signatures in gut microbiota composition and stimulates butyrate production. These effects are associated with changes in innate and acquired immunity.IMPORTANCE The use of a fermented milk product containing the heat-killed probiotic strain Lactobacillus paracasei CBAL74 induces changes in the gut microbiota, promoting the development of butyrate producers. These changes in the gut microbiota composition correlate with increased levels of innate and acquired immunity biomarkers.


Assuntos
Bactérias/isolamento & purificação , Ácido Butírico/metabolismo , Microbioma Gastrointestinal , Lacticaseibacillus paracasei/metabolismo , Probióticos/administração & dosagem , Animais , Bactérias/classificação , Bactérias/genética , Bovinos , Pré-Escolar , Produtos Fermentados do Leite/análise , Produtos Fermentados do Leite/microbiologia , Feminino , Fermentação , Trato Gastrointestinal/microbiologia , Humanos , Lactente , Lacticaseibacillus paracasei/química , Masculino
8.
Mediators Inflamm ; 2017: 8392523, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28246439

RESUMO

Crohn's disease (CD) is a chronic inflammation of the intestinal mucosa, characterized by periods of acute recurrence and remission. Depending on the specific region affected, CD is classified as ileal CD or colonic CD. It is largely accepted that the intestinal microbiota is involved in the onset of the pathology. Indeed, a reduced immune tolerance to components of the intestinal commensal microbiota and inflammation of the intestinal barrier typifies patients with CD. Several studies have shown defective expression of intestinal antimicrobial peptides (AMPs) in patients with CD compared to controls, particularly defensins. A reduction in α-defensins is observed in ileal CD, while ß-defensins are increased in colonic CD. In addition to an immunological basis, the disease is frequently associated with genetic alterations including mutations of NOD2 gene. Several therapeutic strategies to circumvent the dysfunction observed in CD are currently under investigation. These include the use of delivery systems to administer endogenous AMPs and the engineering of peptidomimetics that could ameliorate the severity of CD. In this review, the role defensins play in CD and the strategies aimed at overcoming bacterial resistance will be discussed.


Assuntos
Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Defensinas/metabolismo , Mucosa Intestinal/microbiologia , Microbiota , Animais , Peptídeos Catiônicos Antimicrobianos/química , Doença de Crohn/imunologia , Defensinas/imunologia , Modelos Animais de Doenças , Farmacorresistência Bacteriana , Humanos , Íleo/metabolismo , Íleo/microbiologia , Sistema Imunitário , Inflamação , Mucosa Intestinal/imunologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Camundongos , alfa-Defensinas , beta-Defensinas
9.
Int J Mol Sci ; 18(2)2017 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-28212280

RESUMO

Urinary tract infections (UTIs) and catheter-associated UTIs (CAUTIs) are the principal hospital-acquired infections. Proteus mirabilis is characterized by several virulence factors able to promote adhesion and biofilm formation and ameliorate the colonization of urinary tract and the formation of crystalline biofilms on the abiotic surface of the urinary catheters. Since, to date, the role of P. mirabilis in the etiopathogenesis of different types of urinary tract infections is not well established, in this study we sought to characterize two different clinically isolated strains of P. mirabilis (PM1 and PM2) with distinctive phenotypes and analyzed various virulence factors possibly implicated in the ability to induce UTIs and CAUTIs. In particular, we analyzed motility, biofilm formation both on abiotic and biotic surfaces of PM1 and PM2 and paralleled these parameters with the ability to induce an inflammatory response in an epithelial cell model. Results showed that PM1 displayed major motility and a capacity to form biofilm and was associated with an anti-inflammatory response of host cells. Conversely, PM2 exhibited lack motility and a had slower organization in biofilm but promoted an increase of proinflammatory cytokine expression in infected epithelial cells. Our study provides data useful to start uncovering the pathologic basis of P. mirabilis-associated urinary infections. The evidence of different virulence factors expressed by PM1 and PM2 highlights the possibility to use precise and personalized therapies targeting specific virulence pathways.


Assuntos
Biofilmes/crescimento & desenvolvimento , Imunomodulação , Infecções por Proteus/imunologia , Infecções por Proteus/microbiologia , Proteus mirabilis/fisiologia , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Fenótipo , Proteus mirabilis/classificação , Proteus mirabilis/isolamento & purificação , Infecções Urinárias/imunologia , Infecções Urinárias/microbiologia , Fatores de Virulência
10.
Molecules ; 22(3)2017 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-28272373

RESUMO

Antimicrobial peptides (AMPs) play a pivotal role in the innate immune responses to Helicobacter pylori (Hp) in humans. ß-Defensins, a class of cationic arginine-rich AMPs, are small peptides secreted by immune cells and epithelial cells that exert antimicrobial activity against a broad spectrum of microorganisms, including Gram-positive and Gram-negative bacteria and fungi. During Hp infections, AMP expression is able to eradicate the bacteria, thereby preventing Hp infections in gastrointestinal tract. It is likely that gastric ß-defensins expression is increased during Hp infection. The aim of this review is to focus on increased knowledge of the role of ß-defensins in response to Hp infection. We also briefly discuss the potential use of AMPs, either alone or in combination with conventional antibiotics, for the treatment of Hp infection.


Assuntos
Infecções por Helicobacter/microbiologia , Helicobacter pylori/efeitos dos fármacos , Interações Hospedeiro-Patógeno , beta-Defensinas/farmacologia , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Células Epiteliais/metabolismo , Gastrite/etiologia , Infecções por Helicobacter/complicações , Infecções por Helicobacter/tratamento farmacológico , Helicobacter pylori/patogenicidade , Humanos , Neoplasias Gástricas/etiologia , Relação Estrutura-Atividade , Fatores de Virulência , beta-Defensinas/química , beta-Defensinas/metabolismo , beta-Defensinas/uso terapêutico
11.
Adv Exp Med Biol ; 879: 91-105, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26659265

RESUMO

Lipopolysaccharide (LPS) is one of the principal bacterial products known to elicit inflammation. Cells of myeloid lineage such as monocytes and macrophages, but also epithelial cells give rise to an inflammatory response upon LPS stimulation. This phenomenon implies reprogramming of cell specific gene expression that can occur through different mechanisms including epigenetic modifications. Given their intrinsic nature, epigenetic modifications may be involved both in the acute response to LPS and in the establishment of a preconditioned genomic state (epigenomic memory) that may potentially influence the host response to further contacts with microorganisms. Information has accumulated during the last years aimed at elucidating the epigenetic mechanisms which underlie the cellular LPS response. These findings, summarized in this chapter, will hopefully be a good basis for a definition of the complete cascade of LPS-induced epigenetic events and their biological significance in different cell types.


Assuntos
Bactérias/metabolismo , Infecções Bacterianas/metabolismo , Epigênese Genética , Lipopolissacarídeos/metabolismo , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/patologia , Humanos
12.
Front Cell Neurosci ; 18: 1433747, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39175504

RESUMO

The Aryl hydrocarbon receptor (AHR) is a cytosolic receptor and ligand-activated transcription factor widely expressed across various cell types in the body. Its signaling is vital for host responses at barrier sites, regulating epithelial renewal, barrier integrity, and the activities of several types of immune cells. This makes AHR essential for various cellular responses during aging, especially those governing inflammation and immunity. In this review, we provided an overview of the mechanisms by which the AHR mediates inflammatory response at gut and brain level through signals from intestinal microbes. The age-related reduction of gut microbiota functions is perceived as a trigger of aberrant immune responses linking gut and brain inflammation to neurodegeneration. Thus, we explored gut microbiome impact on the nature and availability of AHR ligands and outcomes for several signaling pathways involved in neurodegenerative diseases and age-associated decline of brain functions, with an insight on Parkinson's and Alzheimer's diseases, the most common neurodegenerative diseases in the elderly. Specifically, we focused on microbial tryptophan catabolism responsible for the production of several AHR ligands. Perspectives for the development of microbiota-based interventions targeting AHR activity are presented for a healthy aging.

13.
Eur J Med Chem ; 264: 115981, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38086192

RESUMO

The occurrence of increased antibiotic resistance has reduced the availability of drugs effective in the control of infectious diseases, especially those caused by various combinations of bacteria and/or fungi that are often associated with poorer patient outcomes. In the hunt for novel antibiotics of interest to treat polymicrobial diseases, molecules bearing guanidine moieties have recently come to the fore in designing and optimizing antimicrobial agents. Due to their remarkable antibacterial and antifungal activities, labdane diterpenes are also attracting increasing interest in antimicrobial drug discovery. In this study, six different guanidines prenylated with labdanic fragments were synthesized and evaluated for their antimicrobial properties. Assays were carried out against both non-resistant and antibiotic-resistant bacteria strains, while their possible antifungal activities have been tested on the yeast Candida albicans. Two of the synthesized compounds, namely labdan-8,13(R)-epoxy-15-oyl guanidine and labdan-8,13(S)-epoxy-15-oyl guanidine, were finally selected as the best candidates for further developments in drug discovery, due to their antimicrobial effects on both Gram-negative and Gram-positive bacterial strains, their fungicide action, and their moderate toxicity in vivo on zebrafish embryos. The study also provides insights into the structure-activity relationships of the guanidine-functionalized labdane-type diterpenoids.


Assuntos
Anti-Infecciosos , Diterpenos , Animais , Humanos , Antifúngicos/farmacologia , Guanidina/farmacologia , Peixe-Zebra , Anti-Infecciosos/farmacologia , Antibacterianos/farmacologia , Bactérias , Diterpenos/farmacologia , Candida albicans , Guanidinas/farmacologia , Testes de Sensibilidade Microbiana
14.
J Med Chem ; 67(16): 14256-14276, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39115219

RESUMO

The widespread and irrational use of azole antifungal agents has led to an increase of azole-resistant Candida albicans strains with an urgent need for combination drug therapy, enhancing the treatment efficacy. Here, we report the discovery of a first-in-class pyrazole-isoxazole, namely, 5b, that showed remarkable growth inhibition against the C. albicans ATCC 10231 strain in combination with voriconazole, acting as a downregulator of ERG 11 (Cyp51) gene expression with a significant reduction of the yeast-to-hypha morphological transition. Furthermore, C. albicans CYP51 enzyme assay and in-depth molecular docking studies unveiled the unique ability of the combination of 5b and voriconazole to completely fill the CYP51 binding sites. In vivo studies using a Galleria mellonella model confirmed the previously in vitro observed synergistic effect of 5b with voriconazole. Also considering its biocompatibility in a cellular model of human keratinocytes, these results indicate that 5b represents a promising compound for a further optimization campaign.


Assuntos
Antifúngicos , Candida albicans , Farmacorresistência Fúngica , Isoxazóis , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Pirazóis , Voriconazol , Antifúngicos/farmacologia , Antifúngicos/química , Voriconazol/farmacologia , Candida albicans/efeitos dos fármacos , Pirazóis/farmacologia , Pirazóis/química , Animais , Humanos , Isoxazóis/farmacologia , Isoxazóis/química , Sinergismo Farmacológico , Mariposas/microbiologia , Mariposas/efeitos dos fármacos , Candidíase/tratamento farmacológico , Candidíase/microbiologia , Modelos Animais de Doenças , Relação Estrutura-Atividade , Azóis/farmacologia , Azóis/química , Azóis/uso terapêutico
15.
Bioorg Med Chem ; 21(22): 7038-46, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24094434

RESUMO

Two new acylated styrylpyrones, one 5-methoxy-1(3H)-isobenzofuranone glucoside and a hydroxymethyl-orcinol derivative, along with sixteen known aromatic metabolites, including lignans, quinic acid derivatives low-molecular weight phenol glucosides, have been isolated from the methanol extract of Helichrysum italicum, a medicinal plant typical of the Mediterranean vegetation. The structures of these compounds have been elucidated on the basis of extensive 2D-NMR spectroscopic analyses, including COSY, TOCSY, HSQC, CIGAR-HMBC, H2BC and HSQC-TOCSY, along with Q-TOF HRMS(2) analysis. Selected compounds were evaluated for their anti-biofilm properties against Pseudomonas aeruginosa.


Assuntos
Biofilmes/efeitos dos fármacos , Helichrysum/química , Extratos Vegetais/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Antibacterianos/química , Antibacterianos/farmacologia , Glucosídeos/química , Glucosídeos/isolamento & purificação , Glucosídeos/farmacologia , Espectroscopia de Ressonância Magnética , Conformação Molecular , Extratos Vegetais/química , Folhas de Planta/química , Plantas Medicinais/química , Resorcinóis/química , Resorcinóis/isolamento & purificação , Resorcinóis/farmacologia
16.
Poult Sci ; 102(2): 102222, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36502562

RESUMO

Helicobacter pullorum is recognized as an emerging food-borne pathogen that may colonize the intestinal tract and the liver of avian species and humans causing several gastrointestinal and liver diseases. However, not all strains are reported to be capable of causing clinical disease, thus making poultry as reservoir for the zoonotic transmission of the infection through carcass contamination of broilers at slaughter. In poultry, the prevalence of this bacterium could be underestimated and the available data mainly refer to conventional rearing systems, whereas free-range and organic breedings have been poorly investigated. Therefore, this study was aimed to characterize the caecal microbiota community of free-range grown chickens and determine the presence and the relative abundance of H. pullorum by using NGS-based 16S rDNA sequencing. A total of 18 chickens reared for 56 d on a semi-extensive management system were euthanized at two time points: 9 birds at 28 d of age (before have access to outdoor; I = Indoor) and other 9 birds at 56 d of age (before slaughter; O = Outdoor). Cecal contents were collected for microbiota analyses. H. pullorum was detected in the cecum of 16/18 samples and its proportion in indoor was significantly higher than outdoor chickens (2.46 and 0.52%, respectively; P < 0.05), showing 78.8% of decrease with the outdoor access of the chickens. Therefore, it may be assumed that the potential for zoonotic infection is less likely. Moreover, H. pullorum was negatively correlated with 17 bacterial species as significantly more abundant in Outdoor microbial caecal communities. Among these, we highlighted the presence of Mucispirillium schaedleri and Oscillospira, already previously associated with a healthy gut and thus representing promising gut bacterial markers for host health. Our findings suggest that alternative production systems with outdoor access, may play a crucial role in the establishment of a healthy gut microbiota, which in turn might prevent colonization of harmful bacteria such as Helicobacter pullorum.


Assuntos
Helicobacter , Microbiota , Humanos , Animais , Galinhas/microbiologia , Helicobacter/genética , Bactérias , Ceco/microbiologia , Microbiota/genética
17.
Sci Rep ; 13(1): 18197, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37875530

RESUMO

The gut-brain axis involves several bidirectional pathway communications including microbiome, bacterial metabolites, neurotransmitters as well as immune system and is perturbed both in brain and in gastrointestinal disorders. Consistently, microbiota-gut-brain axis has been found altered in autism spectrum disorder (ASD). We reasoned that such alterations occurring in ASD may impact both on methylation signatures of human host fecal DNA (HFD) and possibly on the types of human cells shed in the stools from intestinal tract giving origin to HFD. To test this hypothesis, we have performed whole genome methylation analysis of HFD from an age-restricted cohort of young children with ASD (N = 8) and healthy controls (N = 7). In the same cohort we have previously investigated the fecal microbiota composition and here we refined such analysis and searched for eventual associations with data derived from HFD methylome analysis. Our results showed that specific epigenetic signatures in human fecal DNA, especially at genes related to inflammation, associated with the disease. By applying methylation-based deconvolution algorithm, we found that the HFD derived mainly from immune cells and the relative abundance of those differed between patients and controls. Consistently, most of differentially methylated regions fitted with genes involved in inflammatory response. Interestingly, using Horvath epigenetic clock, we found that ASD affected children showed both epigenetic and microbiota age accelerated. We believe that the present unprecedented approach may be useful for the identification of the ASD associated HFD epigenetic signatures and may be potentially extended to other brain disorders and intestinal inflammatory diseases.


Assuntos
Transtorno do Espectro Autista , Microbioma Gastrointestinal , Humanos , Criança , Pré-Escolar , Transtorno do Espectro Autista/metabolismo , Microbioma Gastrointestinal/genética , Disbiose/microbiologia , Metilação de DNA , Inflamação/genética , Inflamação/complicações
18.
Front Nutr ; 10: 1143004, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37599675

RESUMO

Obesity is associated with gastrointestinal (GI) tract and central nervous system (CNS) disorders. High-fat diet (HFD) feeding-induced obesity in mice induces dysbiosis, causing a shift toward bacteria-derived metabolites with detrimental effects on metabolism and inflammation: events often contributing to the onset and progression of both GI and CNS disorders. Palmitoylethanolamide (PEA) is an endogenous lipid mediator with beneficial effects in mouse models of GI and CNS disorders. However, the mechanisms underlining its enteroprotective and neuroprotective effects still need to be fully understood. Here, we aimed to study the effects of PEA on intestinal inflammation and microbiota alterations resulting from lipid overnutrition. Ultramicronized PEA (30 mg/kg/die per os) was administered to HFD-fed mice for 7 weeks starting at the 12th week of HFD regimen. At the termination of the study, the effects of PEA on inflammatory factors and cells, gut microbial features and tryptophan (TRP)-kynurenine metabolism were evaluated. PEA regulates the crosstalk between the host immune system and gut microbiota via rebalancing colonic TRP metabolites. PEA treatment reduced intestinal immune cell recruitment, inflammatory response triggered by HFD feeding, and corticotropin-releasing hormone levels. In particular, PEA modulated HFD-altered TRP metabolism in the colon, rebalancing serotonin (5-HT) turnover and reducing kynurenine levels. These effects were associated with a reshaping of gut microbiota composition through increased butyrate-promoting/producing bacteria, such as Bifidobacterium, Oscillospiraceae and Turicibacter sanguinis, with the latter also described as 5-HT sensor. These data indicate that the rebuilding of gut microbiota following PEA supplementation promotes host 5-HT biosynthesis, which is crucial in regulating intestinal function.

19.
Front Vet Sci ; 9: 904522, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35909674

RESUMO

Characterizing the gut microbiota of free-range and alternative poultry production systems provides information, which can be used to improve poultry welfare, performance, and environmental sustainability. Gut microbiota influence not only the health and metabolism of the host but also the presence of zoonotic agents contaminating food of animal origin. In this study, the composition and diversity of the cecal microbiota community of free-range grown chickens were characterized by 16S rDNA high-throughput Illumina sequencing. Significant differences were observed in the composition of chicken cecal microbiota at the time points of 28 days of age (Indoor group) and 56 days of age (Outdoor group), i.e., before and after the outdoor access period of chicken groups. The Outdoor group showed a richer and more complex microbial community, characterized by the onset of new phyla such as Deferribacterota and Synergistota, while the Indoor group showed an increase in Campylobacterota. At the species level, it is noteworthy that the occurrence of Mucispirillum schaedleri in Outdoor group is known to potentially stimulate mucus layer formation in the distal intestinal tract, thus being associated with a healthy gut. We also report a significant decrease in the Outdoor group of Helicobacter pullorum, highlighting that the lower abundance at the age of slaughter reduced the possibility to contaminate chickens' carcasses and, consequently, its zoonotic potential. As revealed by a mutual exclusion study in network analysis, H. pullorum was present only if Bacteroides barnesiae, an uncultured organism of the genus Synergistes, and Bacteroides gallinaceum were absent. Finally, microbiome predictive analysis revealed an increase of vitamins and micronutrient biosyntheses such as queuosine (Q) and its precursor pre Q0, in the Outdoor group, suggesting that the outdoor evolved microbiota of chickens do contribute to the vitamin pool of the gut and the biosynthesis of micronutrients involved in vital cell processes.

20.
Biomolecules ; 11(2)2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33499115

RESUMO

The bidirectional microbiota-gut-brain axis has raised increasing interest over the past years in the context of health and disease, but there is a lack of information on molecular mechanisms underlying this connection. We hypothesized that change in microbiota composition may affect brain epigenetics leading to long-lasting effects on specific brain gene regulation. To test this hypothesis, we used Zebrafish (Danio Rerio) as a model system. As previously shown, treatment with high doses of probiotics can modulate behavior in Zebrafish, causing significant changes in the expression of some brain-relevant genes, such as BDNF and Tph1A. Using an ultra-deep targeted analysis, we investigated the methylation state of the BDNF and Tph1A promoter region in the brain and gut of probiotic-treated and untreated Zebrafishes. Thanks to the high resolution power of our analysis, we evaluated cell-to-cell methylation differences. At this resolution level, we found slight DNA methylation changes in probiotic-treated samples, likely related to a subgroup of brain and gut cells, and that specific DNA methylation signatures significantly correlated with specific behavioral scores.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Metilação de DNA , Triptofano Hidroxilase/genética , Alelos , Animais , Comportamento Animal , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Biologia Computacional/métodos , Ilhas de CpG , Epigênese Genética , Feminino , Microbioma Gastrointestinal , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Biblioteca Gênica , Lacticaseibacillus rhamnosus , Masculino , Microbiota , Probióticos , Regiões Promotoras Genéticas , Triptofano Hidroxilase/metabolismo , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA