Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Sci Rep ; 14(1): 16059, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992159

ABSTRACT

Cholangiocarcinoma (CCA) is often diagnosed late, leading to incomplete tumor removal, drug resistance and reduced chemotherapy efficacy. Curcumin has the potential for anti-cancer activity through various therapeutic properties and can improve the efficacy of chemotherapy. We aimed to investigate the synergistic effect of a combination of curcumin and gemcitabine against CCA, targeting the LAT2/glutamine pathway. This combination synergistically suppressed proliferation in gemcitabine-resistant CCA cells (KKU-213BGemR). It also resulted in a remarkable degree of CCA cell apoptosis and cell cycle arrest, characterized by a high proportion of cells in the S and G2/M phases. Knockdown of SLC7A8 decreased the expressions of glutaminase and glutamine synthetase, resulting in inhibited cell proliferation and sensitized CCA cells to gemcitabine treatment. Moreover, in vivo experiments showed that a combination curcumin and gemcitabine significantly reduced tumor size, tumor growth rate and LAT2 expression in a gemcitabine-resistant CCA xenograft mouse model. Suppression of tumor progression in an orthotopic CCA hamster model provided strong support for clinical application. In conclusion, curcumin synergistically enhances gemcitabine efficacy against gemcitabine-resistant CCA by induction of apoptosis, partly via inhibiting LAT2/glutamine pathway. This approach may be an alternative strategy for the treatment of gemcitabine-resistant in CCA patients.


Subject(s)
Apoptosis , Cell Proliferation , Cholangiocarcinoma , Curcumin , Deoxycytidine , Drug Resistance, Neoplasm , Drug Synergism , Gemcitabine , Glutamine , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Animals , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Humans , Curcumin/pharmacology , Drug Resistance, Neoplasm/drug effects , Mice , Glutamine/metabolism , Cell Line, Tumor , Apoptosis/drug effects , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Signal Transduction/drug effects , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Glutaminase/metabolism , Glutaminase/antagonists & inhibitors , Male
2.
PLoS One ; 19(5): e0301907, 2024.
Article in English | MEDLINE | ID: mdl-38814931

ABSTRACT

BACKGROUND: Opisthorchis viverrini (O. viverrini, Ov) infection and consumption of high-fat and high-fructose (HFF) diet exacerbate liver and kidney disease. Here, we investigated the effects of a combination of O. viverrini infection and HFF diet on kidney pathology via changes in the gut microbiome and host proteome in hamsters. METHODOLOGY/PRINCIPAL FINDINGS: Twenty animals were divided into four groups; 1) fed a normal diet not infected with O. viverrini (normal group), 2) fed an HFF diet and not infected with O. viverrini (HFF), 3) fed a normal diet and infected with O. viverrini (Ov), and 4) fed an HFF diet and infected with O. viverrini (HFFOv). DNA was extracted from fecal samples and the V3-V4 region of the bacterial 16S rRNA gene sequenced on an Illumina MiSeq sequencing platform. In addition, LC/MS-MS analysis was done. Histopathological studies and biochemical assays were also conducted. The results indicated that the HFFOv group exhibited the most severe kidney injury, manifested as elevated KIM-1 expression and accumulation of fibrosis in kidney tissue. The microbiome of the HFFOv group was more diverse than in the HFF group: there were increased numbers of Ruminococcaceae, Lachnospiraceae, Desulfovibrionaceae and Akkermansiaceae, but fewer Eggerthellaceae. In total, 243 host proteins were identified across all groups. Analysis using STITCH predicted that host proteome changes may lead to leaking of the gut, allowing molecules such as soluble CD14 and p-cresol to pass through to promote kidney disease. In addition, differential expression of TGF-beta-activated kinase 1 and MAP3K7-binding protein 2 (Tab2, involving renal inflammation and injury) are predicted to be associated with kidney disease. CONCLUSIONS/SIGNIFICANCE: The combination of HFF diet and O. viverrini infection may promote kidney injury through alterations in the gut microbiome and host proteome. This knowledge may suggest an effective strategy to prevent kidney disease beyond the early stages.


Subject(s)
Diet, High-Fat , Fructose , Gastrointestinal Microbiome , Metagenomics , Opisthorchiasis , Proteomics , Animals , Opisthorchiasis/complications , Opisthorchiasis/parasitology , Opisthorchiasis/pathology , Opisthorchiasis/metabolism , Diet, High-Fat/adverse effects , Metagenomics/methods , Cricetinae , Proteomics/methods , Kidney Diseases/metabolism , Kidney Diseases/parasitology , Kidney Diseases/microbiology , Kidney Diseases/pathology , Kidney Diseases/etiology , Opisthorchis , Male , Proteome , Kidney/pathology , Kidney/metabolism , Kidney/microbiology , Mesocricetus , RNA, Ribosomal, 16S/genetics
3.
Heliyon ; 10(9): e30104, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38720699

ABSTRACT

Background: Microcystin-leucine arginine (MC-LR) is a cyanobacterial hepatotoxic toxin found in water sources worldwide, including in northeastern Thailand, where opisthorchiasis-associated cholangiocarcinoma (CCA) is most prevalent. MC-LR is a potential carcinogen; however, its involvement in liver fluke-associated CCA remains ambiguous. Here, we aimed to evaluate the effect of MC-LR on the progression of CCA via the Wnt/ß-catenin pathway in vitro. Methods: Cell division, migration, cell cycle transition, and MC-LR transporter expression were evaluated in vitro through MTT assay, wound healing assay, flow cytometry, and immunofluorescence staining, respectively. Following a 24-h treatment of cultured cells with 1, 10, 100, and 1,000 nM of MC-LR, the proliferative effect of MC-LR on the Wnt/ß-catenin signaling pathway was investigated using immunoblotting and qRT-PCR analysis. Immunohistochemistry was used to determine ß-catenin expression in CCA tissue compared to adjacent tissue. Results: Human immortalized cholangiocyte cells (MMNK-1) and a human cell line established from opisthorchiasis-associated CCA (KKU-213B) expressed the MC-LR transporter and internalized MC-LR. Exposure to 10 nM and 100 nM of MC-LR notably enhanced cells division and migration in both cell lines (P < 0.05) and markedly elevated the percentage of S phase cells (P < 0.05). MC-LR elevated PP2A expression by activating the Wnt/ß-catenin signaling pathway and suppressing phosphatase activity. Inhibition of the ß-catenin destruction complex genes (Axin1 and APC) led to the upregulation of ß-catenin and its downstream target genes (Cyclin D1 and c-Jun). Inhibition of Wnt/ß-catenin signaling by MSAB confirmed these results. Additionally, ß-catenin was significantly expressed in cancerous tissue compared to adjacent areas (P < 0.001). Conclusions: Our findings suggest that MC-LR promotes cell proliferation and progression of CCA through Wnt/ß-catenin pathway. Further evaluation using invivo experiments is needed to confirm this observation. This finding could promote health awareness regarding MC-LR intake and risk of CCA.

4.
Am J Trop Med Hyg ; 110(5): 951-952, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38579698

ABSTRACT

Infection with Strongyloides stercoralis is often asymptomatic but can be life-threatening in immunocompromised patients, which can be prevented by ivermectin (IVM) treatment. The efficacy of IVM has been reported to have lessened over time in some regions as a consequence of prolonged use and mass treatment campaigns. Ivermectin has been used in Thailand for more than a decade; therefore, we investigated the efficacy of a single dose (200 µg/kg) of IVM against in asymptomatic strongyloidiasis in northeastern Thailand. Fecal samples were collected before and 2 weeks after treatment and were analyzed for the presence of Strongyloides using a modified agar plate culture and the formalin-ethyl acetate concentration technique. Our results showed that single-dose IVM treatment successfully eliminated S. stercoralis infection in asymptomatic individuals in the endemic area with a 100% cure rate, indicating the high efficacy of IVM treatment in strongyloidiasis in northeast Thailand.


Subject(s)
Feces , Ivermectin , Strongyloides stercoralis , Strongyloidiasis , Ivermectin/therapeutic use , Strongyloidiasis/drug therapy , Humans , Animals , Strongyloides stercoralis/drug effects , Thailand , Feces/parasitology , Male , Female , Adult , Middle Aged , Antiparasitic Agents/therapeutic use , Young Adult , Adolescent , Treatment Outcome
5.
BMC Complement Med Ther ; 24(1): 178, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689275

ABSTRACT

BACKGROUND: Gastrointestinal cancer (GIC) ranks as the highest cause of cancer-related deaths globally. GIC patients are often diagnosed at advanced stages, limiting effective treatment options. Chemotherapy, the common GIC recommendation, has significant disadvantages such as toxicity and adverse effects. Natural products contain substances with diverse pharmacological characteristics that promise for use in cancer therapeutics. In this study, the flower of renowned Asian medicinal plant, Shorea roxburghii was collected and extracted to investigate its phytochemical contents, antioxidant, and anticancer properties on GIC cells. METHODS: The phytochemical contents of Shorea roxburghii extract were assessed using suitable methods. Phenolic content was determined through the Folin-Ciocalteu method, while flavonoids were quantified using the aluminum chloride (AlCl3) method. Antioxidant activity was evaluated using the FRAP and DPPH assays. Cytotoxicity was assessed in GIC cell lines via the MTT assay. Additionally, intracellular ROS levels and apoptosis were examined through flow cytometry techniques. The correlation between GIC cell viability and phytochemicals, 1H-NMR analysis was conducted. RESULTS: Among the four different solvent extracts, ethyl acetate extract had the highest phenolic and flavonoid contents. Water extract exhibited the strongest reducing power and DPPH scavenging activity following by ethyl acetate. Interestingly, ethyl acetate extract demonstrated the highest inhibitory activity against three GIC cell lines (KKU-213B, HepG2, AGS) with IC50 values of 91.60 µg/ml, 39.38 µg/ml, and 35.59 µg/ml, while showing less toxicity to normal fibroblast cells. Ethyl acetate extract induced reactive oxygen species and apoptosis in GIC cell lines by downregulating anti-apoptotic protein Bcl-2. Metabolic profiling-based screening revealed a positive association between reduced GIC cell viability and phytochemicals like cinnamic acid and its derivatives, ferulic acid and coumaric acid. CONCLUSIONS: This study highlights the potential of natural compounds in Shorea roxburghii in the development of more effective and safer anticancer agents as options for GIC as well as shedding light on new avenues for cancer treatment.


Subject(s)
Gastrointestinal Neoplasms , Plant Extracts , Humans , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cell Line, Tumor , Gastrointestinal Neoplasms/drug therapy , Apoptosis/drug effects , Antioxidants/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Survival/drug effects , Metabolomics , Phytochemicals/pharmacology , Flavonoids/pharmacology , Flavonoids/analysis
6.
Sci Rep ; 13(1): 22444, 2023 12 17.
Article in English | MEDLINE | ID: mdl-38105269

ABSTRACT

The microRNA miR-205-5p has diverse effects in different malignancies, including cholangiocarcinoma (CCA), but its effects on CCA progression is unclear. Here we investigated the role and function of miR-205-5p in CCA. Three CCA cell lines and human serum samples were found to have much higher expression levels of miR-205-5p than seen in typical cholangiocyte cell lines and healthy controls. Inhibition of miR-205-5p suppressed CCA cell motility, invasion and proliferation of KKU-213B whereby overexpression of miR-205-5p promoted cell proliferation and motility of KKU-100 cells. Bioinformatics tools (miRDB, TargetScan, miRWalk, and GEPIA) all predicted various miR-205-5p targets. Experiments using miR-205-5p inhibitor and mimic indicated that homeodomain-interacting protein kinase 3 (HIPK3) was a potential direct target of miR-205-5p. Overexpression of HIPK3 using HIPK3 plasmid cloning DNA suppressed migration and proliferation of KKU-100 cells. Notably, HIPK3 expression was lower in human CCA tissues than in normal adjacent tissues. High HIPK3 expression was significantly associated with longer survival time of CCA patients. Multivariate regression analysis indicated tissue HIPK3 levels as an independent prognostic factor for CCA patients. These findings indicate that overexpression of miR-205-5p promotes CCA cells proliferation and migration partly via HIPK3-dependent way. Therefore, targeting miR-205-5p may be a potential treatment approach for CCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , MicroRNAs , Protein Serine-Threonine Kinases , Humans , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cholangiocarcinoma/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Serine-Threonine Kinases/genetics
7.
Heliyon ; 9(9): e19859, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37809389

ABSTRACT

Understanding gut bacterial composition and proteome changes in patients with early-stage chronic kidney disease (CKD) could lead to better methods of controlling the disease progression. Here, we investigated the gut microbiome and microbial functions in patients with S. stercoralis infection (strongyloidiasis) and early-stage CKD. Thirty-five patients with early stages (1-3) of CKD were placed in two groups matched for population characteristics and biochemical parameters, 12 patients with strongyloidiasis in one group and 23 uninfected patients in the other. From every individual, a sample of their feces was obtained and processed for 16S rRNA sequencing and metaproteomic analysis using tandem liquid chromatography-mass spectrometry (LC-MS/MS). Strongyloides stercoralis infection per se did not significantly alter gut microbial diversity. However, certain genera (Bacteroides, Faecalibacterium, Fusicatenibacter, Sarcina, and Anaerostipes) were significantly more abundant in infection-free CKD patients than in infected individuals. The genera Peptoclostridium and Catenibacterium were enriched in infected patients. Among the significantly altered genera, Fusicatenibacter and Anaerostipes were the most correlated with renal parameters. The relative abundance of members of the genus Fusicatenibacter was moderately positively correlated with estimated glomerular filtration rate (eGFR) (r = 0.335, p = 0.049) and negatively with serum creatinine (r = -0.35, p = 0.039). Anaerostipes, on the other hand, showed a near-significant positive correlation with eGFR (r = 0.296, p = 0.084). Individuals with S. stercoralis infection had higher levels of bacterial proteins involved in amino-acid metabolism. Analysis using STITCH predicted that bacterial amino-acid metabolism may also be involved in the production of colon-derived uremic toxin (indole), a toxic substance known to promote CKD. Strongyloides stercoralis infection is, therefore, associated with reduced abundance of Fusicatenibacter and Anaerostipes (two genera possibly beneficial for kidney function) and with increased bacterial amino-acid metabolism in the early-stages of CKD, potentially producing uremic toxin. This study provides useful information for prevention of progression of CKD beyond the early stages.

8.
Microbes Environ ; 38(3)2023.
Article in English | MEDLINE | ID: mdl-37635077

ABSTRACT

Gut dysbiosis and changes in short-chain fatty acids (SCFAs) occur in end-stage chronic kidney disease (CKD); however, the degree of these changes in the gut microbiome and serum SCFA profiles in the early stages of CKD,| |particularly in| |CKD| |of unknown etiology (CKDu), is unclear. We herein investigated the gut microbiome and SCFA profiles of early-stage CKD patients (CKD stages 1-3) in a community in Khon Kaen Province, Thailand. Seventy-two parasite-free participants were distributed among a healthy control group (HC, n=18) and three patient groups (an underlying disease group [UD, n=18], early-stage CKD with underlying disease [CKD-UD, n=18], and early-stage CKD of unknown etiology, [CKDu, n=18]). Fecal DNA was individually extracted and pooled for groups of six individuals (three pools in each group) to examine the composition of the gut microbiome using next-generation sequencing. A SCFA ana-lysis was performed on serum samples from each individual using gas chromatography-mass spectrometry. The results revealed that microbial abundance differed between the healthy group and all patient groups (UD, CKD-UD, and CKDu). [Eubacterium]_coprostanoligenes_group was more abundant in the CKDu group than in the HC and CKD-UD groups. Furthermore, serum concentrations of acetate, a major SCFA component, were significantly lower in all patient groups than in the HC group. The present results indicate that minor changes in the gut microbiome and a significant decrease in serum acetate concentrations occur in early-stage CKDu, which may be important for the development of prevention strategies for CKD patients.


Subject(s)
Gastrointestinal Microbiome , Renal Insufficiency, Chronic , Humans , Chronic Kidney Diseases of Uncertain Etiology , Thailand
10.
Sci Rep ; 13(1): 4216, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36918707

ABSTRACT

We explored the impact of chronic Strongyloides stercoralis infection on the gut microbiome and microbial activity in a longitudinal study. At baseline (time-point T0), 42 fecal samples from matched individuals (21 positive for strongyloidiasis and 21 negative) were subjected to microbiome 16S-rRNA sequencing. Those positive at T0 (untreated then because of COVID19 lockdowns) were retested one year later (T1). Persistent infection in these individuals indicated chronic strongyloidiasis: they were treated with ivermectin and retested four months later (T2). Fecal samples at T1 and T2 were subjected to 16S-rRNA sequencing and LC-MS/MS to determine microbial diversity and proteomes. No significant alteration of indices of gut microbial diversity was found in chronic strongyloidiasis. However, the Ruminococcus torques group was highly over-represented in chronic infection. Metaproteome data revealed enrichment of Ruminococcus torques mucin-degrader enzymes in infection, possibly influencing the ability of the host to expel parasites. Metaproteomics indicated an increase in carbohydrate metabolism and Bacteroidaceae accounted for this change in chronic infection. STITCH interaction networks explored highly expressed microbial proteins before treatment and short-chain fatty acids involved in the synthesis of acetate. In conclusion, our data indicate that chronic S. stercoralis infection increases Ruminococcus torques group and alters the microbial proteome.


Subject(s)
COVID-19 , Strongyloides stercoralis , Strongyloidiasis , Humans , Animals , Strongyloidiasis/parasitology , Proteome , Persistent Infection , Longitudinal Studies , Ruminococcus , Chromatography, Liquid , Communicable Disease Control , Tandem Mass Spectrometry , Feces/parasitology
11.
Sci Rep ; 12(1): 18188, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36307490

ABSTRACT

In Thailand, chronic kidney disease (CKD) screening was reported in 2009 with an overall prevalence of 17.5% and the highest at 22.2% in the northeastern region. This study aimed to find out CKD prevalence of the Kidney Disease Improving Global Outcomes criteria and their related risk factors in the rural community. A population-based study was conducted in the rural sub-districts of northeastern Thailand. Data of socio-demographic status, lifestyle, underlying diseases, blood pressure, and body mass index were recorded. Blood and urine analysis was conducted along with ultrasonography of kidneys. Specimen collection and analyses were repeated after 3 months, and the factors associated with CKD were studied by logistic regression analysis. A total of 2205 participants with a mean age of 57.8 ± 11.7 years and female predominance (66.7%) completed the study. The prevalence of CKD was 26.8%, i.e., stages 1 (7.3%); stage 2 (9.0%); stage 3a (6.0%); stage 3b (2.8%); stage 4 (1.4%); and stage 5 (0.3%). Hypertension, diabetes mellitus, and renal stones were the major underlying diseases. Only 3.5% of the participants were aware of having CKD. An increase in age, male, unemployment, current smoking, diabetes, hypertension, underweight, anemia, hyperuricemia, and leukocytosis were significantly associated factors with the disease. The study revealed that CKD has developed as a significant public health problem in rural northeastern Thailand and one out of every four people has CKD. Therefore, early interventions are essential for the proper management and prevention of CKD.


Subject(s)
Diabetes Mellitus , Hypertension , Renal Insufficiency, Chronic , Male , Female , Humans , Middle Aged , Aged , Prevalence , Thailand/epidemiology , Renal Insufficiency, Chronic/epidemiology , Risk Factors , Hypertension/epidemiology , Diabetes Mellitus/epidemiology
12.
PLoS Negl Trop Dis ; 16(9): e0010302, 2022 09.
Article in English | MEDLINE | ID: mdl-36067216

ABSTRACT

BACKGROUND: Strongyloides stercoralis infection typically causes severe symptoms in immunocompromised patients. This infection can also alter the gut microbiota and is often found in areas where chronic kidney disease (CKD) is common. However, the relationship between S. stercoralis and the gut microbiome in chronic kidney disease (CKD) is not understood fully. Recent studies have shown that gut dysbiosis plays an important role in the progression of CKD. Hence, this study aims to investigate the association of S. stercoralis infection and gut microbiome in CKD patients. METHODOLOGY/PRINCIPAL FINDINGS: Among 838 volunteers from Khon Kaen Province, northeastern Thailand, 40 subjects with CKD were enrolled and divided into two groups (S. stercoralis-infected and -uninfected) matched for age, sex and biochemical parameters. Next-generation technology was used to amplify and sequence the V3-V4 region of the 16S rRNA gene to provide a profile of the gut microbiota. Results revealed that members of the S. stercoralis-infected group had lower gut microbial diversity than was seen in the uninfected group. Interestingly, there was significantly greater representation of some pathogenic bacteria in the S. stercoralis-infected CKD group, including Escherichia-Shigella (P = 0.013), Rothia (P = 0.013) and Aggregatibacter (P = 0.03). There was also a trend towards increased Actinomyces, Streptococcus and Haemophilus (P > 0.05) in this group. On the other hand, the S. stercoralis-infected CKD group had significantly lower representation of SCFA-producing bacteria such as Anaerostipes (P = 0.01), Coprococcus_1 (0.043) and a non-significant decrease of Akkermansia, Eubacterium rectale and Eubacterium hallii (P > 0.05) relative to the uninfected group. Interesting, the genera Escherichia-Shigella and Anaerostipes exhibited opposing trends, which were significantly related to sex, age, infection status and CKD stages. The genus Escherichia-Shigella was significantly more abundant in CKD patients over the age of 65 years and infected with S. stercoralis. A correlation analysis showed inverse moderate correlation between the abundance of the genus of Escherichia-Shigella and the level of estimated glomerular filtration rate (eGFR). CONCLUSIONS/SIGNIFICANCE: Conclusion, the results suggest that S. stercoralis infection induced gut dysbiosis in the CKD patients, which might be involved in CKD progression.


Subject(s)
Renal Insufficiency, Chronic , Strongyloides stercoralis , Strongyloidiasis , Aged , Animals , Bacteria/genetics , Dysbiosis/microbiology , Feces/microbiology , Humans , RNA, Ribosomal, 16S/genetics , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/microbiology , Strongyloides stercoralis/genetics , Strongyloidiasis/complications , Thailand
13.
Biosci Microbiota Food Health ; 41(3): 121-129, 2022.
Article in English | MEDLINE | ID: mdl-35854692

ABSTRACT

Intestinal parasitic infections can change gut microbiota and short-chain fatty acids (SCFAs). We aimed to study the interaction among Strongyloides stercoralis, human gut microbiota, and serum SCFAs in a community. Fifty-two subjects in Donchang sub-district, Khon Kaen Province, northeastern Thailand, were included based on specific inclusion and exclusion criteria. Characteristics of the participants were matched between those positive for S. stercoralis infection alone (no other intestinal parasites; Ss+, n=26) and uninfected controls (infection status confirmed by polymerase chain reaction (PCR); Ss-, n=26). Serum short-chain fatty acids were evaluated by gas chromatography-mass spectrometry. DNA was extracted from individual faecal samples and then pooled into two groups (Ss+ and Ss-) for amplification and sequencing of the V3-V4 region of the 16S gene with next-generation technology. We explored the impact of infection with S. stercoralis on the faecal microbiota: individuals infected with this parasite exhibited increased alpha diversity of bacteria. At the genus level, gut microbiota in Ss+ patients showed high abundances of Escherichia-Shigella and Bacteroides but low abundances of the genera Bifidobacterium, Lactobacillus, and Blautia. PCR of individual samples to identify certain species of interest gave results consistent with those from next-generation sequencing of pooled samples and showed that significantly more Ss+ samples contained Bacteroides fragilis. Intriguingly, a major SCFA, acetic acid, was significantly decreased in S. stercoralis infection. In conclusion, S. stercoralis infection caused an imbalance of gut microbiota and decreased acetic acid in serum. This information adds to the knowledge concerning the effect of intestinal nematode-related chronic diseases.

14.
J Nutr Biochem ; 107: 109053, 2022 09.
Article in English | MEDLINE | ID: mdl-35643287

ABSTRACT

A combination of Opisthorchis viverrini infection and high fat/high fructose diets (HFa/HFr) intake is likely to enhance fatty liver and kidney pathologies. Here we investigated the combined effects of chronic O. viverrini infection and HFa/HFr intake on liver and kidney pathologies, metabolism, and gut microbiome in hamsters. Animals were infected with O. viverrini and fed with either standard chow (OV group) or HFa/HFr diet (OH group) and non-infected hamsters were fed with either standard chow (NC) or HFa/HFr diet (HF) for 8 months. The OH group exhibited dyslipidemia and the highest severity of fatty liver. Tubular damage, inflammatory cell infiltration, and tubular fibrosis were the most prominently observed in this group, supported by increased expression of KIM-1, HMGB-1, and MCP-1. Urinary 1H NMR metabolic profiles revealed that tauro-ß-muricholic acid level was increased in the OV and OH groups, whereas metabolites involved in the TCA cycle and gut microbiota-associated metabolites (phenylacetylglycine, trimethylamine, and trimethylamine-N-oxide) were lower in OV, HF and OH groups compared to the NC group. Gut microbial profiles of the OH group were also different from other groups. In conclusion, O. viverrini infection and HFa/HFr diet-induced disturbance of metabolites and gut microbiota associated with concurrent liver and kidney pathologies in hamsters.


Subject(s)
Fatty Liver , Opisthorchiasis , Opisthorchis , Animals , Cricetinae , Fatty Liver/metabolism , Fructose/metabolism , Kidney/pathology , Liver/metabolism , Opisthorchiasis/complications , Opisthorchiasis/metabolism , Opisthorchiasis/pathology
15.
Molecules ; 27(3)2022 Feb 06.
Article in English | MEDLINE | ID: mdl-35164357

ABSTRACT

Proteus mirabilis is a significant cause of urinary tract infection that may contribute to struvite stones. Anti-infection of this bacterium and anti-struvite formation must be considered. Sida acuta Burm. F. (SA) has been used for the treatment of diseases related to kidneys. Therefore, we investigated the effects of the SA leaf ethanolic extract (SAEE) on growth and on virulent factors (swarming motility and urease activity) of Proteusmirabilis isolated from kidney stone formers. We also evaluated anti-struvite crystal formation and phytochemical constituents of SAEE. The minimum inhibitory concentrations (MICs) of SAEE against three clinical P. mirabilis isolates were 8 mg/mL. Intriguingly, the 1/2MIC of SAEE had significant inhibitory effects on the swarming motility and urease activity of clinical P. mirabilis isolates when compared with the condition without SAEE. The SAEE at the various concentrations significantly inhibited the average weights of struvite crystals in a dose-dependent manner, compared with the control. The phytochemical analysis revealed that SAEE contained catechin, chlorogenic acid, rutin, and ferulic acid. This study indicated that SAEE has anti-P. mirabilis and anti-struvite crystal activities via its bioactive compounds. For this reason, SAEE may be developed as a new agent for the treatment of struvite stone induced by P. mirabilis.


Subject(s)
Phytochemicals/analysis , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Proteus mirabilis/drug effects , Sida Plant/chemistry , Struvite/chemistry , Humans
16.
World J Microbiol Biotechnol ; 37(11): 194, 2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34642828

ABSTRACT

The emergence of drug-resistant tuberculosis is a major global public health threat. Thailand is one of the top 14 countries with high tuberculosis and multi-drug resistant tuberculosis rates. Immediate detection of drug-resistant tuberculosis is necessary to reduce mortality and morbidity by effectively providing treatment to ameliorate the formation of resistant strains. Limited data exist of mutation profiles in Northeastern Thailand. Here, 65 drug-resistant Mycobacterium tuberculosis isolates were used to detect mutations by polymerase chain reaction (PCR) and DNA sequencing. In the katG gene, mutations were occurred in 47 (79.7%) among 59 isoniazid resistant samples. For rpoB gene, 31 (96.9%) were observed as mutations in 32 rifampicin resistant isolates. Of 47 katG mutation samples, 45 (95.7%) had mutations in katG315 codon and 2 (4.3%) showed novel mutations at katG365 with amino acid substitution of CCG-CGG (Pro-Arg). Moreover, out of 31 rpoB mutation isolates, the codon positions rpoB516, rpoB526, rpoB531 and rpoB533 were 3 (9.7%), 8 (25.8%), 11 (35.5%) and 1 (3.2%), respectively. Seven isolates of double point mutation were found [rpoB516, 526; 1 (3.2%) and rpoB516, 531; 6 (19.4%)]. In addition, 1 (3.2%) sample had triple point mutation at codon positions rpoB516, 526 and 531. Common and novel mutation codons of the rpoB and katG genes were generated. Although DNA sequencing showed high accuracy, conventional PCR could be applied as an initial marker for screening drug-resistant Mycobacterium tuberculosis isolates in limit resources region. Mutations reported here should be considered when developing new molecular diagnostic methods for implementation in Northeastern Thailand.


Subject(s)
Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Tuberculosis, Multidrug-Resistant/microbiology , Tuberculosis/microbiology , Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Catalase/genetics , DNA, Bacterial/genetics , DNA-Directed RNA Polymerases/genetics , Drug Resistance, Bacterial , Drug Resistance, Multiple, Bacterial , Humans , Isoniazid/pharmacology , Mutation , Mycobacterium tuberculosis/isolation & purification , Polymerase Chain Reaction , Sequence Analysis, DNA , Thailand/epidemiology , Tuberculosis/epidemiology , Tuberculosis, Multidrug-Resistant/epidemiology
17.
Am J Trop Med Hyg ; 105(4): 1117-1123, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34339389

ABSTRACT

Ivermectin (IVM) is a widely used anthelmintic. However, with widespread use comes the risk of the emergence of IVM resistance, particularly in strongyloidiasis. Adenosine triphosphate (ATP)-binding cassette (ABC) transporter genes play an important role in the IVM-resistance mechanism. Here, we aimed to establish an animal experimental model of IVM resistance by frequent treatment of Strongyloides ratti with subtherapeutic doses of IVM, resistance being evaluated by the expression levels of ABC transporter genes. Rats infected with S. ratti were placed in experimental groups as follows: 1) untreated control (control); 2) treated with the mutagen ethyl methanesulfonate (EMS); 3) injected with 100 µg/kg body weight of IVM (IVM); 4) treated with a combination of EMS and IVM (IVM+EMS). Parasites were evaluated after four generations. Extent of IVM resistance was assessed using IVM sensitivity, larval development, and expression of ABC genes. By the F4 generation, S. ratti in the IVM group exhibited significantly higher levels of IVM resistance than did other groups according to in vitro drug-sensitivity tests and inhibition of larval development (IC50 = 36.60 ng/mL; 95% CI: 31.6, 42.01). Expression levels of ABC isoform genes (ABCA, ABCF, and ABCG) were statistically significantly higher in the IVM-resistant line compared with the susceptible line. In conclusion, IVM subtherapeutic doses induced IVM resistance in S. ratti by the F4 generation with corresponding upregulation of some ABC isoform genes. The study provides a model for inducing and assessing drug resistance in Strongyloides.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Drug Resistance , Gene Expression Regulation/drug effects , Ivermectin/administration & dosage , Ivermectin/pharmacology , Strongyloides ratti/drug effects , ATP-Binding Cassette Transporters/genetics , Animals , Antiparasitic Agents/administration & dosage , Antiparasitic Agents/pharmacology , Drug Administration Schedule , Male , Rats , Up-Regulation
18.
Toxicol Rep ; 8: 1346-1357, 2021.
Article in English | MEDLINE | ID: mdl-34277359

ABSTRACT

We recently developed a modified solid dispersion of curcumin-loaded nanocomplexes (CNCs) in gums which promoted the prolonged and sustained release of curcumin. However, its safety assessment has not yet been investigated. Here, acute and chronic toxicities of CNCs were assayed using mice and hamsters. CNCs were orally administered to the animals. Doses of CNCs used for acute toxicity testing were 0.1, 1.1, 11.0 g/kg body weight for mice and 0.2, 2.1 and 21.4 g/kg body weight for hamsters. Doses of CNCs for chronic toxicity testing were 0.09, 0.27, 0.8 g/kg body weight/day for mice and 0.18, 0.54 and 1.61 g/kg body weight/day for hamsters. This regimen was followed daily for 6 months. Low and medium doses of CNCs did not induce any side effects in acute and chronic toxicity tests in either animal species. However, in acute toxicity testing, the organ-weight to body-weight ratio of spleen was significantly increased in mice treated with 11 g/kg body weight along with elevated levels of some biochemical parameters. There was a significant increase in organ-weight to body-weight ratios of stomach, liver and heart in hamsters treated with 21.4 g/kg body weight, but no elevated levels of biochemical parameters. Oral LD50 of CNCs in mice and hamsters were 8.9 and 16.8 g/kg body weight (equivalent to 2.5 and 4.7 g curcumin/kg body weight), respectively. Daily CNCs high-dose treatment for 6 months significantly increased organ-weight to body-weight ratios of stomach and intestine in mice and of lung and heart in hamsters. Elevated levels of glucose, total protein, ALT, AST and globulin in mice, and increased levels of AST, but decrease in cholesterol, in hamsters were concurrently observed with inflammation in liver and lung. These abnormalities were resolved within 28 days after cessation of treatment. The no-observed-adverse-effect level of CNCs was determined at 0.27 and 0.54 g/kg body weight/day in mice and hamsters. In conclusion, toxicity of high-dose CNCs treatment was graded as very low, possibly due to the components of the nanocomplex.

19.
J Proteome Res ; 20(8): 3940-3951, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34270897

ABSTRACT

Opisthorchis viverrini (Ov) infection causes hepatobiliary diseases and is a major risk factor for cholangiocarcinoma. While several omics approaches have been employed to understand the pathogenesis of opisthorchiasis, effects of Ov infection on the host systemic metabolism and fecal microbiota have not been fully explored. Here, we used a 1H NMR spectroscopy-based metabolic phenotyping approach to investigate Ov infection-induced metabolic disturbances at both the acute (1 month postinfection, 1 mpi) and chronic (4 mpi) stages in hamsters. A total of 22, 3, and 4 metabolites were found to be significantly different in the liver, serum, and urine, respectively, between Ov+ and Ov- groups. Elevated levels of hepatic amino acids and tricarboxylic acid (TCA)-cycle intermediates (fumarate and malate) were co-observed with liver injury in acute infection, whereas fibrosis-associated metabolites (e.g., glycine and glutamate) increased at the chronic infection stage. Lower levels of lipid signals ((CH2)n and CH2CH2CO) and higher levels of lysine and scyllo-inositol were observed in serum from Ov+ hamsters at 1 mpi compared to Ov- controls. Urinary levels of phenylacetylglycine (a host-bacterial cometabolite) and tauro-ß-muricholic acid were higher in the Ov+ group, which coexisted with hepatic and mild kidney fibrosis. Furthermore, Ov+ animals showed higher relative abundances of fecal Methanobrevibacter (Archaea), Akkermansia, and Burkholderia-Paraburkholderia compared to the noninfected controls. In conclusion, along with liver and kidney pathologies, O. viverrini infection resulted in hepatic and mild renal pathologies, disturbed hepatic amino acid metabolism and the TCA cycle, and induced changes in the fecal microbial composition and urinary host-microbial cometabolism. This study provides the initial step toward an understanding of local and systemic metabolic responses of the host to O. viverrini infection.


Subject(s)
Bile Duct Neoplasms , Opisthorchiasis , Opisthorchis , Animals , Bile Ducts, Intrahepatic , Cricetinae , Kidney , Liver , Opisthorchiasis/complications
SELECTION OF CITATIONS
SEARCH DETAIL