Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Biomed Pharmacother ; 178: 117283, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39126775

RESUMO

Rifaximin, derived from rifamycin, is a broad-spectrum antibiotic by inhibiting bacterial RNA synthesis. Rifaximin has a very low intestinal absorption and exerts its antimicrobial activity primarily in the intestinal tract. It regulates the gut microbiota with limited side effects systemically. Rifaximin has been recommended for the treatment of hepatic encephalopathy but some studies shed light on its medicinal effects in many other diseases. For instance, rifaximin may suppress the progression of liver fibrosis and its related complications, and ameliorate metabolic dysfunction-associated steatotic liver disease and alcohol-associated liver disease, etc. Rifaximin can also mediate anti-inflammation, antiproliferation, and proapoptotic events by activating pregnane X receptor, which is efficious in cancers such as colon cancer. In addition, some investigations have shown rifaximin may play a therapeutic role in various autoimmune and neurological disorders. However, these findings still need more real-world practices and in-depth investigations to obtain more precise indications and fully elucidate the multifaceted potentials of rifaximin.


Assuntos
Hepatopatias , Rifaximina , Rifaximina/uso terapêutico , Rifaximina/farmacologia , Humanos , Hepatopatias/tratamento farmacológico , Hepatopatias/microbiologia , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Rifamicinas/uso terapêutico , Rifamicinas/farmacologia
2.
Sci Rep ; 14(1): 18140, 2024 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-39103611

RESUMO

Rifaximin is FDA-approved for treatment of irritable bowel syndrome with diarrhea (IBS-D), but poor solubility may limit its efficacy against microbes in the mucus layer, e.g. Escherichia coli. Here we evaluate adding the mucolytic N-acetylcysteine (NAC) to improve rifaximin efficacy. In a resazurin checkerboard assay, combining rifaximin with NAC had significant synergistic effects in reducing E. coli levels. The optimal rifaximin + NAC combination was then tested in a validated rat model of IBS-D (induced by cytolethal distending toxin [CdtB] inoculation). Rats were inoculated with vehicle and treated with placebo (Control-PBS) or rifaximin + NAC (Control-Rif + NAC, safety), or inoculated with CdtB and treated with placebo (CdtB-PBS), rifaximin (CdtB-Rifaximin), or rifaximin + NAC (CdtB-Rif + NAC) for 10 days. CdtB-inoculated rats (CdtB-PBS) developed wide variability in stool consistency (P = 0.0014) vs. controls (Control-PBS). Stool variability normalized in rats treated with rifaximin + NAC (CdtB-Rif + NAC) but not rifaximin alone (CdtB-Rifaximin). Small bowel bacterial levels were elevated in CdtB-PBS rats but normalized in CdtB-Rif + NAC but not CdtB-Rifaximin rats. E. coli and Desulfovibrio spp levels (each associated with different IBS-D microtypes) were also elevated in CdtB-inoculated (CdtB-PBS) but normalized in CdtB-Rif + NAC rats. Cytokine levels normalized only in CdtB-Rif + NAC rats, in a manner predicted to be associated with reduced diarrhea driven by reduced E. coli. These findings suggest that combining rifaximin with NAC may improve the percentage of IBS-D patients responding to treatment.


Assuntos
Acetilcisteína , Diarreia , Modelos Animais de Doenças , Escherichia coli , Síndrome do Intestino Irritável , Rifaximina , Animais , Rifaximina/farmacologia , Rifaximina/uso terapêutico , Acetilcisteína/farmacologia , Acetilcisteína/administração & dosagem , Ratos , Escherichia coli/efeitos dos fármacos , Diarreia/tratamento farmacológico , Diarreia/microbiologia , Síndrome do Intestino Irritável/tratamento farmacológico , Síndrome do Intestino Irritável/microbiologia , Masculino , Ratos Sprague-Dawley , Quimioterapia Combinada
3.
Brain Behav ; 14(7): e3621, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38970239

RESUMO

INTRODUCTION: Hepatic encephalopathy (HE) is a severe neuropsychiatric complication of liver diseases characterized by neuroinflammation. The efficacies of nonabsorbable rifaximin (RIF) and lactulose (LAC) have been well documented in the treatment of HE. [18F]PBR146 is a translocator protein (TSPO) radiotracer used for in vivo neuroinflammation imaging. This study investigated anti-neuroinflammation effect of RIF or/and LAC in chronic HE rats by [18F]PBR146 micro-PET/CT. METHODS: Bile duct ligation (BDL) operation induced chronic HE models, and this study included Sham+normal saline (NS), BDL+NS, BDL+RIF, BDL+LAC, and BDL+RIF+LAC groups. Behavioral assessment was performed to analyze the motor function, and fecal samples were collected after successfully established the chronic HE model (more than 28 days post-surgery). In addition, fecal samples collection and micro-PET/CT scans were performed sequentially. And we also collected the blood plasma, liver, intestinal, and brain samples after sacrificing the rats for further biochemical and pathological analyses. RESULTS: The RIF- and/or LAC-treated BDL rats showed similar behavioral results with Sham+NS group, while the treatment could not reverse the biliary obstruction resulting in sustained liver injury. The RIF or/and LAC treatments can inhibit IFN-γ and IL-10 productions. The global brain uptake values of [18F]PBR146 in BDL+NS group was significantly higher than other groups (p < .0001). The brain regions analysis showed that the basal ganglia, hippocampus, and cingulate cortex had radiotracer uptake differences among groups (all p < .05), which were consistent with the brain immunohistochemistry results. Sham+NS group was mainly enriched in Christensenella, Coprobacillus, and Pseudoflavonifractor. BDL+NS group was mainly enriched in Barnesiella, Alloprevotella, Enterococcus, and Enterorhabdus. BDL+RIF+LAC group was enriched in Parabacteroides, Bacteroides, Allobaculum, Bifidobacterium, and Parasutterella. CONCLUSIONS: RIF or/and LAC had anti-neuroinflammation in BDL-induced chronic HE rats with gut microbiota alterations. The [18F]PBR146 could be used for monitoring RIF or/and LAC treatment efficacy of chronic HE rats.


Assuntos
Encefalopatia Hepática , Lactulose , Ratos Sprague-Dawley , Rifaximina , Animais , Encefalopatia Hepática/tratamento farmacológico , Encefalopatia Hepática/diagnóstico por imagem , Encefalopatia Hepática/metabolismo , Rifaximina/farmacologia , Ratos , Masculino , Lactulose/farmacologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Modelos Animais de Doenças , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/diagnóstico por imagem , Fármacos Gastrointestinais/farmacologia , Fármacos Gastrointestinais/administração & dosagem , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Radioisótopos de Flúor , Proteínas de Transporte , Receptores de GABA-A
4.
Int J Biol Macromol ; 275(Pt 1): 133582, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38955301

RESUMO

Inulin as a natural polysaccharide regulates intestinal microorganisms, and improves the immune and gastrointestinal function. In order to explore the effect of inulin on pulmonary metastasis of colon cancer, we set up a CT26 injected pulmonary metastatic model. The results showed that inulin used alone did not improve pulmonary metastasis of colon cancer, while inulin combined with rifaximin significantly prolonged the survival time of mice, and inhibited pulmonary metastasis compared with model and inulin groups. Inulin treatment increased the abundance of harmful bacteria such as Proteobacteria and Actinobacteria, while combined treatment decreased their abundance and increased the abundance of beneficial bacteria containing Firmicutes and Eubacterium which belonged to the bile acid-related bacteria. The combination treatment decreased the content of primary bile acids and secondary bile acids in the feces of mice, especial for DCA and LCA which were the agonists of TGR5. Furthermore, the combination treatment reduced the mRNA expression of the TGR5, cyclin dependent kinase 4, cyclin 1 and CDK2, increased the mRNA expression of p21 in the lung, down-regulated the level of NF-κB p65, and up-regulated the level of TNF-α compared with the model group. The above may be the reason for the better use of the combination treatment.


Assuntos
Ácidos e Sais Biliares , Neoplasias do Colo , Inulina , Neoplasias Pulmonares , Rifaximina , Inulina/farmacologia , Animais , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Neoplasias do Colo/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Ácidos e Sais Biliares/metabolismo , Rifaximina/farmacologia , Rifaximina/uso terapêutico , Microbioma Gastrointestinal/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Linhagem Celular Tumoral , Masculino , Camundongos Endogâmicos BALB C
5.
Biomater Adv ; 161: 213862, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38678666

RESUMO

The emergence of antibiotic resistance makes the treatment of bacterial infections difficult and necessitates the development of alternative strategies. Targeted drug delivery systems are attracting great interest in overcoming the limitations of traditional antibiotics. Here, we aimed for targeted delivery of rifaximin (RFX) by decorating RFX-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) with synthetic P6.2 peptide, which was used as a targeting agent for the first time. Our results showed that encapsulation of RFX into NPs increased its antibacterial activity by improving its solubility and providing controlled release, while P6.2 modification allowed targeting of NPs to S. aureus bacterial cells. A promising therapeutic approach for bacterial infections, these P6.2-conjugated RFX-loaded PLGA NPs (TR-NP) demonstrated potent antibacterial activity against both strains of S. aureus. The antibacterial activity of RFX-loaded PLGA NPs (R-NP) showed significant results with an increase of 8 and 16-fold compared to free RFX against S. aureus and MRSA, respectively. Moreover, the activity of targeted nanoparticles was found to be increased 32 or 16-fold with an MBC value of 0.0078 µg/mL. All nanoparticles were found to be biocompatible at doses where they showed antimicrobial activity. Finally, it revealed that P6.2-conjugated targeted nanoparticles extremely accumulated in S. aureus rather than E. coli.


Assuntos
Antibacterianos , Nanopartículas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Rifaximina , Infecções Estafilocócicas , Staphylococcus aureus , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Rifaximina/farmacologia , Rifaximina/química , Nanopartículas/química , Staphylococcus aureus/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/administração & dosagem , Infecções Estafilocócicas/tratamento farmacológico , Testes de Sensibilidade Microbiana , Humanos , Rifamicinas/farmacologia , Rifamicinas/química , Rifamicinas/administração & dosagem , Animais , Sistemas de Liberação de Medicamentos/métodos , Portadores de Fármacos/química
6.
Brain Behav Immun ; 119: 363-380, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38608741

RESUMO

The gut microbiota is altered in epilepsy and is emerging as a potential target for new therapies. We studied the effects of rifaximin, a gastrointestinal tract-specific antibiotic, on seizures and neuropathology and on alterations in the gut and its microbiota in a mouse model of temporal lobe epilepsy (TLE). Epilepsy was induced by intra-amygdala kainate injection causing status epilepticus (SE) in C57Bl6 adult male mice. Sham mice were injected with vehicle. Two cohorts of SE mice were fed a rifaximin-supplemented diet for 21 days, starting either at 24 h post-SE (early disease stage) or at day 51 post-SE (chronic disease stage). Corresponding groups of SE mice (one each disease stage) were fed a standard (control) diet. Cortical ECoG recording was done at each disease stage (24/7) for 21 days in all SE mice to measure the number and duration of spontaneous seizures during either rifaximin treatment or control diet. Then, epileptic mice ± rifaximin and respective sham mice were sacrificed and brain, gut and feces collected. Biospecimens were used for: (i) quantitative histological analysis of the gut structural and cellular components; (ii) markers of gut inflammation and intestinal barrier integrity by RTqPCR; (iii) 16S rRNA metagenomics analysis in feces. Hippocampal neuronal cell loss was assessed in epileptic mice killed in the early disease phase. Rifaximin administered for 21 days post-SE (early disease stage) reduced seizure duration (p < 0.01) and prevented hilar mossy cells loss in the hippocampus compared to epileptic mice fed a control diet. Epileptic mice fed a control diet showed a reduction of both villus height and villus height/crypt depth ratio (p < 0.01) and a decreased number of goblet cells (p < 0.01) in the duodenum, as well as increased macrophage (Iba1)-immunostaining in the jejunum (p < 0.05), compared to respective sham mice. Rifaximin's effect on seizures was associated with a reversal of gut structural and cellular changes, except for goblet cells which remained reduced. Seizure duration in epileptic mice was negatively correlated with the number of mossy cells (p < 0.01) and with villus height/crypt depth ratio (p < 0.05). Rifaximin-treated epileptic mice also showed increased tight junctions (occludin and ZO-1, p < 0.01) and decreased TNF mRNA expression (p < 0.01) in the duodenum compared to epileptic mice fed a control diet. Rifaximin administered for 21 days in chronic epileptic mice (chronic disease stage) did not change the number or duration of seizures compared to epileptic mice fed a control diet. Chronic epileptic mice fed a control diet showed an increased crypt depth (p < 0.05) and reduced villus height/crypt depth ratio (p < 0.01) compared to respective sham mice. Rifaximin treatment did not affect these intestinal changes. At both disease stages, rifaximin modified α- and ß-diversity in epileptic and sham mice compared to respective mice fed a control diet. The microbiota composition in epileptic mice, as well as the effects of rifaximin at the phylum, family and genus levels, depended on the stage of the disease. During the early disease phase, the abundance of specific taxa was positively correlated with seizure duration in epileptic mice. In conclusion, gut-related alterations reflecting a dysfunctional state, occur during epilepsy development in a TLE mouse model. A short-term treatment with rifaximin during the early phase of the disease, reduced seizure duration and neuropathology, and reversed some intestinal changes, strengthening the therapeutic effects of gut-based therapies in epilepsy.


Assuntos
Modelos Animais de Doenças , Epilepsia do Lobo Temporal , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Rifaximina , Convulsões , Animais , Rifaximina/uso terapêutico , Rifaximina/farmacologia , Camundongos , Masculino , Microbioma Gastrointestinal/efeitos dos fármacos , Convulsões/tratamento farmacológico , Epilepsia do Lobo Temporal/tratamento farmacológico , Estado Epiléptico/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Epilepsia/tratamento farmacológico
7.
Metabolism ; 151: 155740, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37995805

RESUMO

BACKGROUND & AIMS: Dysbiosis contributes to alcohol-associated liver disease (ALD); however, the precise mechanisms remain elusive. Given the critical role of the gut microbiota in ammonia production, we herein aim to investigate whether and how gut-derived ammonia contributes to ALD. METHODS: Blood samples were collected from human subjects with/without alcohol drinking. Mice were exposed to the Lieber-DeCarli isocaloric control or ethanol-containing diets with and without rifaximin (a nonabsorbable antibiotic clinically used for lowering gut ammonia production) supplementation for five weeks. Both in vitro (NH4Cl exposure of AML12 hepatocytes) and in vivo (urease administration for 5 days in mice) hyperammonemia models were employed. RNA sequencing and fecal amplicon sequencing were performed. Ammonia and triglyceride concentrations were measured. The gene and protein expression of enzymes involved in multiple pathways were measured. RESULTS: Chronic alcohol consumption causes hyperammonemia in both mice and human subjects. In healthy livers and hepatocytes, ammonia exposure upregulates the expression of urea cycle genes, elevates hepatic de novo lipogenesis (DNL), and increases fat accumulation. Intriguingly, ammonia promotes ethanol catabolism and acetyl-CoA formation, which, together with ammonia, synergistically facilitates intracellular fat accumulation in hepatocytes. Mechanistic investigations uncovered that ATF4 activation, as a result of ER stress induction and general control nonderepressible 2 activation, plays a central role in ammonia-provoked DNL elevation. Rifaximin ameliorates ALD pathologies in mice, concomitant with blunted hepatic ER stress induction, ATF4 activation, and DNL activation. CONCLUSIONS: An overproduction of ammonia by gut microbiota, synergistically interacting with ethanol, is a significant contributor to ALD pathologies.


Assuntos
Amônia , Fígado Gorduroso , Hiperamonemia , Hepatopatias Alcoólicas , Animais , Humanos , Camundongos , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Amônia/efeitos adversos , Amônia/metabolismo , Etanol/efeitos adversos , Etanol/metabolismo , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Hiperamonemia/complicações , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Lipogênese , Fígado/metabolismo , Hepatopatias Alcoólicas/metabolismo , Camundongos Endogâmicos C57BL , Rifaximina/farmacologia
8.
J Microbiol Immunol Infect ; 57(2): 320-327, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38135646

RESUMO

BACKGROUND: The clinical burden of Clostridioides difficile infections (CDIs) remains substantial globally. This study aimed to investigate the ribotypes (RTs) and antimicrobial susceptibility of C. difficile isolates collected in Taiwan. METHODS: C. difficile isolates were prospectively collected from four medical centers in Taiwan from 2019 to 2021. In a reference laboratory, in vitro susceptibility to clindamycin, moxifloxacin, metronidazole, vancomycin, fidaxomicin, and rifaximin were tested, and ribotyping was conducted to determine their genetic diversity. RESULTS: A total of 568 C. difficile isolates were included. Metronidazole resistance was not observed, and the susceptibility rate of vancomycin was 99.5 %. Clindamycin showed poor activity against these isolates, with a resistance rate of 74.8 %. Fidaxomicin exhibited potent activity and 97.4 % of isolates were inhibited at 0.25 µg/mL. Rifaximin MIC90 increased from 0.015 µg/mL in 2019 to 0.03 µg/mL in 2020 and 2021. Of 40 RTs identified, two predominant RTs were RT 078/126 (78, 14 %) and 014/020 (76, 13 %). RT 017, traditional harboring truncated tcdA, accounted for 3 % (20 isolates) and there was no isolate belonging to RT 027. The proportions of RT 078 increased from 11.2 % in 2019 to 17.1 % in 2021, and the predominance of RT 078/126 was more evident in central Taiwan. CONCLUSIONS: Vancomycin, fidaxomicin, and metronidazole remain in vitro effective against clinical C. difficile isolates in Taiwan. The reservoirs and genetic relatedness of two major RTs with zoonotic potentials, RT 078/126 and 014/020, warrant further investigations.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Humanos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Fidaxomicina , Vancomicina/farmacologia , Metronidazol/farmacologia , Ribotipagem , Clindamicina , Rifaximina/farmacologia , Taiwan/epidemiologia , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/epidemiologia , Testes de Sensibilidade Microbiana
9.
Polim Med ; 53(2): 97-103, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37470308

RESUMO

BACKGROUND: The adhesion of Pseudomonas aeruginosa to biotic and abiotic surfaces is responsible for the persistence and development of bacterial infection. OBJECTIVES: To fill the gap in the knowledge regarding the relationship between rifaximin susceptibility and biofilm formation, and to investigate the effect of subinhibitory doses of rifaximin on the adhesion and biofilm formation. MATERIAL AND METHODS: A total of 10 isolates of P. aeruginosa were obtained from 110 urine samples of urinary tract infection (UTI) patients. Biofilm formation on polystyrene microtiter plates, minimum inhibitory concentrations (MICs) of rifaximin against the 10 isolates of P. aeruginosa (Pa1-Pa10), the effect of sub-MICs of rifaximin (0.5 × MIC, 0.25 × MIC, 0.125 × MIC, and 0.06 × MIC) on biofilm formation by the Pa4 isolate to polystyrene microtiter plates, and the adhesion to human epithelial cells (HECs) in vitro were evaluated. RESULTS: The MICs of rifaximin against 10 isolates ranged from 62.5 µg/mL to 1000 µg/mL. The Pa4 isolate produced the highest level of biofilm formation, while the MIC of Pa4 was 125 µg/mL. There was no correlation between bacterial susceptibility to rifaximin and biofilm formation (r: -0.016; p > 0.05). Sub-MIC doses of rifaximin significantly reduced the biofilm formation on abiotic surfaces, while only 0.5 × MIC, 0.25 × MIC and 0.12 × MIC of rifaximin reduced the adhesion to HECs significantly (p < 0.05) in a dose-dependent manner. CONCLUSIONS: This pioneering study demonstrated the negative effect of sub-MIC doses of rifaximin on biofilm formation and adhesion to abiotic and biotic surfaces in vitro.


Assuntos
Antibacterianos , Infecções por Pseudomonas , Humanos , Antibacterianos/farmacologia , Rifaximina/farmacologia , Pseudomonas aeruginosa , Biofilmes , Poliestirenos/farmacologia , Infecções por Pseudomonas/microbiologia , Testes de Sensibilidade Microbiana
10.
Microbiol Spectr ; 11(3): e0377722, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37125917

RESUMO

Clostridioides difficile, the primary cause of nosocomial antibiotic-associated diarrhea, has a complex relationship with antibiotics. While the use of broad-spectrum antibiotics disrupts the gut microbiota and increases the risk of C. difficile infection (CDI), antibiotics are also the primary treatment for CDI. However, only a few antibiotics, including vancomycin, fidaxomicin, and rifaximin, are effective against CDI, and resistance to these antibiotics has emerged recently. In this study, we report the identification of two RT027 C. difficile clinical isolates (TGH35 and TGH64) obtained from symptomatic CDI-diagnosed patients in Tampa, Florida in 2016. These two strains showed an elevated minimum inhibitory concentration (MIC) of vancomycin (MIC = 4 µg/mL, compared to the EUCAST breakpoint of 2 µg/mL) and contained a vanRCd 343A>G mutation resulting in a Thr115Ala substitution in the VanRCd response regulator. This mutation was absent in the vancomycin-sensitive control epidemic strain RT027/R20291. TGH64 was also resistant to rifaximin (MIC ≥ 128 µg/mL) and carried the previously reported Arg505Lys and Ile548Met mutations in RpoB. Furthermore, we report on the antimicrobial resistance (AMR) and genomic characterization of additional C. difficile isolates, including RT106/TGH120, RT017/TGH33, and RT017/TGH51, obtained from the same patient sample cohort representing the highly prevalent and regionally distributed C. difficile ribotypes worldwide. Considering that the VanRCd Thr115Ala mutation was also independently reported in seven C. difficile clinical isolates from Texas and Israel in 2019, we recommend epidemiological surveillance to better understand the impact of this mutation on vancomycin resistance. IMPORTANCE The perpetually evolving antimicrobial resistance (AMR) of C. difficile is an important contributor to its epidemiology and is a grave concern to global public health. This exacerbates the challenge of treating the infections caused by this multidrug-resistant causative organism of potentially life-threatening diarrhea. Further, the novel resistance-determining factors can be transferred between different strains and species of bacteria and cause the spread of AMR in clinical, environmental, and community settings. In this study, we have identified a mutation (vanRCd 343A>G) that causes a Thr115Ala substitution and is linked to an increased MIC of vancomycin in clinical isolates of C. difficile obtained from Florida in 2016. Understanding the mechanisms of AMR, especially those of newly evolving strains, is essential to effectively guide antibiotic stewardship policies to combat antibiotic resistance as well as to discover novel therapeutic targets.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Humanos , Vancomicina/farmacologia , Vancomicina/uso terapêutico , Cádmio/farmacologia , Cádmio/uso terapêutico , Rifaximina/farmacologia , Clostridioides , Florida , Infecções por Clostridium/microbiologia , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Testes de Sensibilidade Microbiana , Diarreia/tratamento farmacológico
11.
BMC Microbiol ; 23(1): 22, 2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36658488

RESUMO

BACKGROUND: Rifaximin has been increasingly applied in irritable bowel syndrome (IBS) treatment. Whether there were differences in the effects of rifaximin on microbiota from different intestinal segments, especially the small intestine where rifaximin predominantly acted, has not been confirmed. METHODS: In this study, we used Trichinella spiralis infection to induce post infectious irritable bowel syndrome (PI-IBS) and measured visceral sensitivity of mice by means of abdominal withdrawal reflex (AWR) tests to colorectal distention (CRD). We compared the effects of rifaximin on the composition of ileal, colonic mucosal and fecal microbiota in PI-IBS mice. RESULTS: Rifaximin significantly reduced AWR scores and increased pain threshold in PI-IBS mice, and this effect was associated with the change in the relative abundance of ileal mucosal microbiota. Rifaximin could obviously decrease ileum mucosal microbiota alpha diversity assessed by Shannon microbial diversity index. Meanwhile, the analysis of beta diversity and relative abundance of microbiota at phylum, family and genus levels showed that rifaximin could improve the microbiota structure of ileal mucosa. However, for colonic mucosal and fecal microbiota, this effect of rifaximin was not obvious. Rifaximin could reshape the correlation of genera between different intestinal segments. CONCLUSION: Rifaximin improved visceral hypersensitivity in PI-IBS mice. Rifaximin mainly affected ileal mucosal microbiota, and its improvement effect on IBS might be closely related to the improvement of ileal microbiota structure.


Assuntos
Síndrome do Intestino Irritável , Microbiota , Camundongos , Animais , Síndrome do Intestino Irritável/tratamento farmacológico , Rifaximina/farmacologia , Intestinos , Mucosa Intestinal
12.
J Dairy Sci ; 106(1): 452-461, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36333135

RESUMO

Bovine mastitis is the most commonly diagnosed disease of dairy cows worldwide and causes extensive economic losses to milk producers. Intramammary infection status before dry-off plays a decisive role with respect to udder health and milk yield in the subsequent lactation. The aim of this study was to compare the effect of antibiotic dry cow therapy (DCT) versus no treatment at dry-off on milk yield, somatic cell count (SCC), inflammation of the mammary gland (IMG), and the incidence of clinical mastitis in the subsequent lactation. Dairy herd data from 251 Austrian dairy farms were recorded over an observation period of 12 mo and subsequently analyzed. The data set included 5,018 dairy cows: 2,078 were treated with antibiotics (abDCT group) and 2,940 were not treated (noDCT group) at dry-off. The abDCT group was subdivided, based on the antimicrobial active substances used for drying off, into 4 different groups (penicillins, cloxacillin, cephalosporins, and rifaximin). Based on bacteriological culture results, infections were grouped into those caused by major, minor, and other pathogens. Additionally, the IMG was defined via SCC from milk recording data using a cutoff of 200,000 cells/mL before drying off and after calving. The incidence of clinical mastitis cases within 30 and 90 d in milk was calculated using veterinary diagnosis data. To investigate the effect of different dry cow therapies on the following parameters: milk yield, SCC, and diagnosed clinical mastitis cases, different linear mixed models were constructed. Overall, the abDCT group was determined to have a significantly higher milk yield over 305 d in milk in the subsequent lactation (increase of 6.18%), compared with the noDCT group (increase of 4.29%). Both groups (abDCT and noDCT) demonstrated a decrease in the first SCC after calving compared with the SCC before dry-off, although the treated cows had a significantly higher reduction. Regarding the different antibiotic groups, with exception of the rifaximin treated cows, all antibiotic groups showed a significant difference from not treated cows with respect to SCC. Additionally, we were able to demonstrate that cows with IMG before dry-off had a 2.073 times higher chance of an increased SCC (>200,000 cells/mL) after calving. With respect to the veterinary diagnosis data, neither the IMG before drying off nor the type of DCT had a significant influence on the probability of developing clinical mastitis within 30 or 90 d in milk. Only a small number of treatments was accompanied with a bacteriological examination before drying off. However, the existing data in this study indicates that the intramammary infection status before dry-off in combination with different dry cow treatments influences udder health and milk yield after calving. Nevertheless, further studies with larger data sets of bacteriological examinations are necessary to enable a more in-depth investigation into the effects of different antibiotic substances used for DCT.


Assuntos
Doenças dos Bovinos , Mastite Bovina , Animais , Bovinos , Feminino , Antibacterianos/farmacologia , Doenças dos Bovinos/tratamento farmacológico , Contagem de Células/veterinária , Lactação , Glândulas Mamárias Animais , Mastite Bovina/tratamento farmacológico , Mastite Bovina/epidemiologia , Leite , Estudos Retrospectivos , Rifaximina/farmacologia , Rifaximina/uso terapêutico , Áustria
13.
Cells ; 11(21)2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36359864

RESUMO

Patients with Parkinson's disease (PD) exhibit distinct gut microbiota, which may promote gut-derived inflammation. Rifaximin is a nonabsorbable antibiotic that can modify gut microbiota. The present study investigated the effect of rifaximin on gut microbiota and inflammation status in PD. The study examined the effect of long-term rifaximin treatment on in vivo transgenic PD mice (MitoPark) and short-term rifaximin treatment on patients with PD. Rifaximin treatment caused a significant change in gut microbiota in the transgenic PD mice; in particular, it reduced the relative abundance of Prevotellaceae UCG-001 and increased the relative abundance of Bacteroides, Muribaculum, and Lachnospiraceae UCG-001. Rifaximin treatment attenuated serum interleukin-1ß, interleukin-6 and tumor necrosis factor-α, claudin-5 and occludin, which indicated the reduction of systemic inflammation and the protection of the blood-brain barrier integrity. The rifaximin-treated MitoPark mice exhibited better motor and memory performance than did the control mice, with lower microglial activation and increased neuronal survival in the hippocampus. In the patients with PD, 7-day rifaximin treatment caused an increase in the relative abundance of Flavonifractor 6 months after treatment, and the change in plasma proinflammatory cytokine levels was negatively associated with the baseline plasma interleukin-1α level. In conclusion, the present study demonstrated that rifaximin exerted a neuroprotective effect on the transgenic PD mice by modulating gut microbiota. We observed that patients with higher baseline inflammation possibly benefited from rifaximin treatment. With consideration for the tolerability and safety of rifaximin, randomized controlled trials should investigate the disease-modification effect of long-term treatment on select patients with PD.


Assuntos
Microbioma Gastrointestinal , Doença de Parkinson , Camundongos , Animais , Rifaximina/farmacologia , Rifaximina/uso terapêutico , Microbioma Gastrointestinal/fisiologia , Doença de Parkinson/tratamento farmacológico , Inflamação/tratamento farmacológico , Antibacterianos/farmacologia
14.
J Neurochem ; 163(5): 406-418, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36189686

RESUMO

Circadian rhythm disruption (CRD) is a potential risk factor for developing Alzheimer's disease (AD). However, the mechanistic link between CRD and AD is still not fully understood. CRD may lead to intestinal barrier impairment. Several studies in animals and humans suggest a connection between gut microbiota disturbance, intestinal barrier damage and neurodegenerative diseases. In this study, we investigated the effect of CRD on cognition in mice and explored the role of intestinal barrier and inflammatory responses in this process. CRD modulates the composition of gut microbiota, impairs intestinal barrier integrity, and induces both peripheral and central inflammation and cognitive impairment in mice. Rifaximin, a non-absorbable antibiotic which modulates the gut microbial composition and increases intestinal barrier integrity, effectively suppresses inflammatory responses, and rescues cognitive impairment induced by CRD. Furthermore, the impairment in hippocampal neurogenesis, tau hyperphosphorylation, and loss in synaptic proteins in CRD mice is also reversed by Rifaximin. These data identify that the impaired intestinal barrier integrity related to gut microbiota disturbance plays a key role in CRD-induced inflammatory responses and cognitive impairments in mice, and Rifaximin is effective in preventing CRD-induced cognitive deficit through protecting the gut barrier and ameliorating neuroinflammation.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Microbioma Gastrointestinal , Camundongos , Animais , Humanos , Rifaximina/farmacologia , Ritmo Circadiano , Doenças Neuroinflamatórias , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/prevenção & controle
15.
Medicina (Kaunas) ; 58(9)2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36143954

RESUMO

Background and Objectives: To investigate the long-term efficacy of rifaximin (RFX) for hyperammonemia and efficacy for refractory ascites in patients with cirrhosis. Materials and Methods: We enrolled 112 patients with liver cirrhosis who were orally administered RFX in this study. Changes in the clinical data of patients were evaluated up to 36 months after RFX administration. The primary endpoint was a change in blood ammonia levels. Secondary endpoints included changes in clinical symptoms, Child−Pugh (CP) score, number of hospitalizations, degree of refractory ascites, adverse events, and the relationship between RFX administration and the renin-angiotensin-aldosterone system. Results: An improved rate of overt hepatic encephalopathy (HE) of 82.7% was observed 3 months after RFX administration, which significantly induced a progressive decrease in blood ammonia concentration and an improved CP score up to 36 months. No serious RFX treatment-related adverse events were observed. 36.5% in patients after RFX administration improved refractory ascites. After RFX administration, patients with satisfactory control of hepatic ascites without addition of diuretic had lower renin concentration than those with poor control (p < 0.01). At less than 41 pg/mL renin concentration, the control of refractory ascites was significantly satisfactory (p < 0.0001). Conclusions: RFX reduced blood ammonia concentration and improved hepatic spare ability and the quality of life of patients with long-term HE to up to 36 months. Our study revealed the effects of RFX against refractory ascites, suggesting that renin concentration may be a predictive marker for assessing ascites control.


Assuntos
Encefalopatia Hepática , Amônia , Ascite/complicações , Ascite/etiologia , Diuréticos , Encefalopatia Hepática/complicações , Encefalopatia Hepática/tratamento farmacológico , Humanos , Cirrose Hepática/complicações , Cirrose Hepática/tratamento farmacológico , Qualidade de Vida , Renina , Rifaximina/farmacologia , Rifaximina/uso terapêutico
16.
Molecules ; 27(13)2022 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-35807317

RESUMO

Testicular dysfunction is caused by chronic exposure to environmental pollution, such as malathion, which causes oxidative stress, promoting cell damage. Autophagy is a key cellular process for eliminating malfunctioning organelles, such as the mitochondria (mitophagy), an eminent source of reactive oxygen species (ROS). Autophagy is crucial for protection against testicular damage. Rifaximin (RFX) is a non-absorbable antibiotic that can reshape the gut microbiome, making it effective in different gastrointestinal disorders. Interestingly, the gut microbiome produces short chain fatty acids (SCFAs) in the circulation, which act as signal molecules to regulate the autophagy. In this study, we investigated the regulatory effects of RFX on gut microbiota and its circulating metabolites SCFA and linked them with the autophagy in testicular tissues in response to malathion administration. Moreover, we divided the groups of rats that used malathion and RFX into a two-week group to investigate the mitophagy process and a four-week group to study mitochondriogenesis. The current study revealed that after two weeks of cotreatment with RFX, apoptosis was inhibited, oxidative stress was improved, and autophagy was induced. More specifically, PINK1 was overexpressed, identifying mitophagy activation. After four weeks of cotreatment with RFX, there was an increase in acetate and propionate-producing microflora, as well as the circulating levels of SCFAs. In accordance with this, the expression of PGC-1α, a downstream to SCFAs action on their receptors, was activated. PGC-1α is an upstream activator of mitophagy and mitochondriogenesis. In this sense, the protein expression of TFAM, which regulates the mitochondrial genome, was upregulated along with a significant decrease in apoptosis and oxidative stress. Conclusion: we found that RFX has a positive regulatory effect on mitophagy and mitochondria biogenesis, which could explain the novel role played by RFX in preventing the adverse effects of malathion on testicular tissue.


Assuntos
Microbioma Gastrointestinal , Mitofagia , Animais , Ácidos Graxos Voláteis , Malation/toxicidade , Mitofagia/genética , Estresse Oxidativo/genética , Ratos , Espécies Reativas de Oxigênio/metabolismo , Rifaximina/farmacologia
17.
Reprod Toxicol ; 111: 194-203, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35714934

RESUMO

Although the use of medication during pregnancy is common, information on exposure to the developing fetus and potential teratogenic effects is often lacking. This study used a rat model to examine the placental transfer of three small-molecule drugs with molecular weights ranging from approximately 300 to 800 Da with different physicochemical properties. Time-mated Sprague Dawley (Hsd:SD) rats aged 11-13 weeks were administered either glyburide, rifaximin, or fentanyl at gestational day 15. Maternal blood, placentae, and fetuses were collected at 5 min, 30 min, 1 h, 4 h, 8 h, 24 h, 48 h, and 96 h post-dose. To characterize the rate and extent of placental drug transfer, we calculated several pharmacokinetic parameters such as maximum concentration (Cmax), time to maximum concentration (Tmax), area under the concentration-time curve (AUC), half-life (t1/2), clearance (CL), and volume of distribution (Vd) for plasma, placenta, and fetus tissues. The results indicated showed that fetal exposure was lowest for glyburide, accounting for only 2.2 % of maternal plasma exposure as measured by their corresponding AUC ratio, followed by rifaximin (37.9 %) and fentanyl (172.4 %). The fetus/placenta AUC ratios were found to be 10.7 % for glyburide, 11.8 % for rifaximin, and 39.1 % for fentanyl. These findings suggest that although the placenta acts as a protective shield for the fetus, the extent of protection varies for different drugs and depends on factors such as molecular weight, lipid solubility, transporter-mediated efflux, and binding to maternal and fetal plasma proteins.


Assuntos
Troca Materno-Fetal , Placenta , Animais , Feminino , Fentanila/metabolismo , Fentanila/farmacologia , Feto , Glibureto/metabolismo , Glibureto/farmacologia , Placenta/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Rifaximina/farmacologia
18.
Dig Liver Dis ; 54(10): 1392-1402, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35514019

RESUMO

BACKGROUND: Although gut-derived lipopolysaccharide (LPS) affects the progression of non-alcoholic steatohepatitis (NASH) pathogenesis, few studies have focused on this relationship to develop treatments for NASH. AIMS: To explore the effects of combination with rifaximin and lubiprostone on NASH liver fibrosis through the modulation of gut barrier function. METHODS: To induce steatohepatitis, F344 rats were fed a choline-deficient l-amino acid-defined (CDAA) diet for 12 weeks and received oral administration of rifaximin and/or lubiprostone. Histological, molecular, and fecal microbial analyses were performed. Barrier function in Caco-2 cells were assessed by in vitro assays. RESULTS: Combination rifaximin/lubiprostone treatment significantly suppressed macrophage expansion, proinflammatory responses, and liver fibrosis in CDAA-fed rats by blocking hepatic translocation of LPS and activation of toll-like receptor 4 signaling. Rifaximin and lubiprostone improved intestinal permeability via restoring tight junction proteins (TJPs) with the intestinal activation of pregnane X receptor and chloride channel-2, respectively. Moreover, this combination increased the abundance of Bacteroides, Lactobacillus, and Faecalibacterium as well as decreased that of Veillonella resulting in an increase of fecal short-chain fatty acids and a decrease of intestinal sialidase activity. Both agents also directly suppressed the LPS-induced barrier dysfunction and depletion of TJPs in Caco-2 cells. CONCLUSION: The combination of rifaximin and lubiprostone may provide a novel strategy for treating NASH-related fibrosis.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Acetamidas , Aminoácidos/metabolismo , Aminoácidos/farmacologia , Animais , Células CACO-2 , Canais de Cloreto/metabolismo , Canais de Cloreto/farmacologia , Colina/metabolismo , Colina/farmacologia , Dieta , Humanos , Lipopolissacarídeos/metabolismo , Fígado/patologia , Cirrose Hepática/complicações , Cirrose Hepática/prevenção & controle , Lubiprostona/farmacologia , Neuraminidase/metabolismo , Neuraminidase/farmacologia , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Receptor de Pregnano X/metabolismo , Ratos , Ratos Endogâmicos F344 , Rifaximina/farmacologia , Proteínas de Junções Íntimas/metabolismo , Receptor 4 Toll-Like/metabolismo
19.
Sci Rep ; 12(1): 3789, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35260705

RESUMO

Current knowledge suggests that infection by carbapenem-resistant enterobacteria is preceded by gut colonization. It is hypothesized that colonization is eradicated by non-absorbable antibiotics like rifaximin. We investigated the effect of rifaximin against carbapenem-resistant Klebsiella pneumoniae (CRKP) in vitro and in a mouse model. We studied the in vitro efficacy of rifaximin against 257 CRKP clinical isolates, 188 KPC producers and 69 OXA-48 producers, by minimum inhibitory concentration and time-kill assays. We then developed a model of gut colonization by feeding 30 C57Bl6 mice with 108 cfu of one KPC-KP isolate for 7 days; mice were pre-treated orally with saline, omeprazole or ampicillin. Then, another 60 mice with established KPC-2 gut colonization received orally for 7 consecutive days rifaximin 180 mg/kg dissolved in ethanol and 4% bile or vehicle. On days 0, 3 and 7 stool samples were collected; mice were sacrificed for determination of tissue outgrowth. At a concentration of 1000 µg/ml rifaximin inhibited 84.8% of CRKP isolates. Α 3 × log10 decrease of the starting inoculum was achieved by 100, 250 and 500 µg/ml of rifaximin after 24 h against 25, 55 and 55% of isolates. Pre-treatment with ampicillin was necessary for gut colonization by KPC-KP. Treatment with rifaximin succeeded in reducing KPC-KP load in stool and in the intestine. Rifaximin inhibits at clinically meaningful gut concentrations the majority of CRKP isolates and is efficient against gut colonization by KPC-KP.


Assuntos
Enterobacteriáceas Resistentes a Carbapenêmicos , Infecções por Klebsiella , Ampicilina/farmacologia , Ampicilina/uso terapêutico , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Proteínas de Bactérias/farmacologia , Carbapenêmicos/farmacologia , Modelos Animais de Doenças , Infecções por Klebsiella/tratamento farmacológico , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Rifaximina/farmacologia , Rifaximina/uso terapêutico , beta-Lactamases/farmacologia
20.
J Biosci Bioeng ; 133(6): 567-578, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35339353

RESUMO

Synergistic therapy against the resurgence of bacterial pathogenesis is a modern trend for antibacterial chemotherapy. The phytochemical allicin, found in garlic extract is a commendable antimicrobial agent that can be used in synergistic combination with modern antibiotics. Determination of optimal antibacterial combination for the target species is vital for maximizing efficacy, lowering toxicity, total eradication of the bacterial cells and minimization of the risk of resistance generation. In this present investigation, Hill function-based pharmacodynamics models were employed to elaborate various time-kill kinetics parameters. The bactericidal potency of the synergistic combinations of allicin and individual antibiotic was assessed in comparison to their monotherapy application viz. using sole allicin and sole antibiotics (levofloxacin, ciprofloxacin, oxytetracycline, rifaximin, ornidazole and azithromycin) on actively growing Bacillus subtilis and Escherichia coli bacteria. Here, all the synergistic combinations showed significantly better (t-test p-value < 0.05) killing effect and biofilm reduction potential compared to their respective monotherapy application, where the highest killing effect was observed with rifaximin-allicin combination (kill rate was more than 5.5 h-1). Moreover, the average inhibition potential to protein denaturation by the synergistic combination group was significantly higher (3.4 fold) than the sole antibiotic's group manifests reduction in the dose-related toxicity. The potential of synergism between antibiotics and allicin combination demonstrated greater killing efficiency at significantly lower concentration compared to monotherapy with increased kill rates in all cases.


Assuntos
Antibacterianos , Ácidos Sulfínicos , Antibacterianos/química , Antibacterianos/farmacologia , Bactérias , Dissulfetos , Sinergismo Farmacológico , Escherichia coli , Cinética , Testes de Sensibilidade Microbiana , Rifaximina/farmacologia , Ácidos Sulfínicos/farmacologia , Ácidos Sulfínicos/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...