Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Endocrinol (Lausanne) ; 13: 895970, 2022.
Article in English | MEDLINE | ID: mdl-36093087

ABSTRACT

Background and Aims: Oral xerostomia remains one of the most common complications of differentiated thyroid carcinoma patients (DTC) after radioiodine therapy (RAI). Environmental factors in the etiology of xerostomia are largely unknown. We aimed to characterize the oral microbiota signatures and related biological functions associated with xerostomia and identify environmental factors affecting them. Methods: Saliva was collected from 30 DTC patients with xerostomia (XAs), 32 patients without xerostomia (indicated as non-XAs) following RAI after total thyroidectomy, and 40 healthy people (HCs) for 16S rRNA sequencing analysis. Results: The oral microbiota of XAs and non-XAs exhibited significant differences in α and ß diversities and bacterial taxa. The abundance of porphyromonas, fusobacterium, and treponema_2 were significantly higher in XAs, while the abundance of the streptococcus was lower in the microbiota of non-XAs. Fusobacterium, and porphyromonas were negatively correlated with unstimulated/stimulated whole salivary secretion (USW)/(SWS), while fusobacterium, porphyromonas, and treponema_2 genera levels were positively associated with cumulative radioiodine dose. PICRUSt2 and BugBase suggested a significant difference in the expression of potentially_pathogenic, anaerobic, gram_negative, the arachidonic acid metabolism, and lipopolysaccharide (LPS) biosynthesis between XAs and non-XAs, possibly interdependent on radioiodine-induced inflammation. NetShift analysis revealed that porphyromonas genus might play as a key driver during the process of xerostomia. Five genera effectively distinguished XAs from non-XAs (AUC = 0.87). Conclusion: Our study suggests for the first time that DTC patients with xerostomia after RAI display microbiota profiles and associated functional changes that may promote a pro-inflammatory environment. Dysbiosis of the oral microbiota may contribute to exacerbating the severity of xerostomia. Our results provide a research direction of the interaction mechanism between oral microbiota alteration and the progress of xerostomia.


Subject(s)
Adenocarcinoma , Microbiota , Thyroid Neoplasms , Xerostomia , Humans , Iodine Radioisotopes/adverse effects , RNA, Ribosomal, 16S/genetics , Thyroid Neoplasms/radiotherapy , Xerostomia/etiology
2.
Front Endocrinol (Lausanne) ; 13: 834674, 2022.
Article in English | MEDLINE | ID: mdl-35350100

ABSTRACT

Background: Thyroid hormone withdrawal (THW) in postoperative thyroid cancer patients who need always accompanied by complications (e.g., dyslipidemia and constipation). At present, there are no effective and safe means to alleviate these complications. Purpose: We aimed to assess the oral-gut microbiota profiles in THW patients then investigate whether probiotics could alleviating alleviate THW related complications and investigate whether these therapeutic effects were associated with the oral-gut microbiota state. Methods: Fifty eligible thyroid carcinoma patients undergoing thyroidectomy were randomly assigned to receive probiotics or placebo during THW. Complications were assessed through validated questionnaires and plasma lipid indicators. The complex probiotics preparation was composed of Bifidobacterium infantis, Lactobacillus acidophilus, Enterococcus faecalis, and Bacillus cereus. Results: Probiotics alleviated lack of energy, constipation, weight gain, and dry mouth and decreased the levels of fecal/serum LPS and plasma lipid indicators (total cholesterol, triglycerides, low-density lipoprotein, and apolipoprotein A) (P < 0.05). Gut and oral microbial diversity were significantly decreased after THW, while an increased microbial dysbiosis index (MDI) was observed. Probiotics distinctly restored the gut and oral microbial diversity. Increased Holdemanella, Enterococcus, and Coprococcus_2, while decreased Fusobacterium, Eubacterium_ruminantium_group, Ruminococcus_1, and Parasutterella in the gut were found after probiotics intervention. Lack of energy, constipation, weight gain, and dyslipidemia were seen to be related to the above microbiota. In addition, probiotics reduced oral Prevotella_9, Haemophilus, Fusobacterium, and Lautropia, which were positively correlated with the occurrence of dry mouth. Conclusion: Probiotics reduce the incidence of complications in patients after THW, which may be related to modifying the oral and gut microbiota. Clinical Trial Registration: [https://clinicaltrials.gov/], identifier America Clinical Trial Registry NCT03574051.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Probiotics , Thyroid Neoplasms , Dysbiosis/etiology , Dysbiosis/therapy , Humans , Iodine Radioisotopes , Probiotics/therapeutic use , Thyroid Hormones/pharmacology , Thyroid Neoplasms/surgery , Thyroidectomy/adverse effects
3.
Front Oncol ; 10: 526009, 2020.
Article in English | MEDLINE | ID: mdl-33324541

ABSTRACT

BACKGROUND: Ileus and postoperative ileus (POI) are common complications of colorectal cancer (CRC). However, little is known about the gut microbiota associated with ileus. METHOD: Differences in gut microbiota were evaluated by 16S rRNA gene sequencing. We characterized the gut microbiota in 85 CRC patients (cohort 1) and detected differences, and an independent cohort composed of 38 CRC patients (cohort 2) was used to evaluate the results. RESULTS: The gut microbiota of CRC patients with and without ileus exhibited large differences in alpha- and beta-diversities and bacterial taxa. The Firmicutes-to-Bacteroidetes ratio and microbial dysbiosis index (MDI) showed greater dysbiosis among ileus patients than among those without ileus. According to the location of CRC, the difference in gut microbiota between patients with and without ileus was more obvious in those with distal CRC than in those with proximal CRC. Finally, Faecalibacterium was significantly reduced in the postoperative perioperative period in patients with ileus. Thus, we used Faecalibacterium as a biomarker for predicting perioperative or POI: the AUC value was 0.74 for perioperative ileus and 0.67 for POI that appeared at 6 months after hospital discharge. The predictive power was evaluated in Cohort 2, with an AUC value of 0.79. CONCLUSION: These findings regarding difference of gut microbiota in postoperative CRC patients may provide a theoretical basis for the use of microbiota as biomarkers for the prediction of POI.

4.
Front Microbiol ; 11: 1106, 2020.
Article in English | MEDLINE | ID: mdl-32595614

ABSTRACT

Evaluating the risk of colorectal metachronous adenoma (MA), which is a precancerous lesion, is necessary for metachronous colorectal cancer (CRC) precaution among CRC patients who had underwent surgical removal of their primary tumor. Here, discovery cohort (n = 41) and validation cohort (n = 45) of CRC patients were prospectively enrolled in this study. Mucosal and fecal samples were used for gut microbiota analysis by sequencing the 16S rRNA genes. Significant reduction of microbial diversity was noted in MA (P < 0.001). A signature defined by decreased abundance of eight genera and increased abundance of two genera strongly correlated with MA. The microbiota-based random forest (RF) model, established utilizing Escherichia-Shigella, Acinetobacter together with BMI in combination, achieved AUC values of 0.885 and 0.832 for MA, predicting in discovery and validation cohort, respectively. The RF model was performed as well for fecal and tumor adjacent mucosal samples with an AUC of 0.835 and 0.889, respectively. Gut microbiota profile of MA still existed in post-operative cohort patients, but the RF model could not be performed well on this cohort, with an AUC of 0.61. Finally, we introduced a risk score based on Escherichia-Shigella, Acinetobacter and BMI, and synchronous-adenoma achieved AUC values of 0.94 and 0.835 in discovery and validation cohort, respectively. This study presented a comprehensive landscape of gut microbiota in MA, demonstrated that the gut microbiota-based models and scoring system achieved good ability to predict the risk for developing MA after surgical resection. Our study suggests that gut microbiota is a potential predictive biomarker for MA.

5.
Mol Ther Oncolytics ; 16: 262-271, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32181327

ABSTRACT

This study aimed to assess the effectiveness of inhibiting cholesterol acyltransferase 1 (ACAT-1) in chimeric antigen receptor T (CAR-T) cells on potentiating the antitumor response against mesothelin (MSLN)-expressing pancreatic carcinoma (PC) cells. We engineered ACAT-1-inhibited CAR-T cells (CAR-T-1847 and CAR-T-1848) using the targeting MSLN CAR lentiviral vector and small interfering RNA (siRNA) targeting the conserved region of the ACAT-1 gene, and characterized the efficacy of these modified CAR-T cells in terms of the cytotoxicity and cytokine release of both MSLN-positive and MSLN-negative PC cells using in vitro methods and in vivo mouse xenografts. The ACAT-1-inhibited CAR-T-1847 and CAR-T-1848 cells showed a higher cytotoxicity at effector-to-target cell (E:T) ratios of 8:1 and 10:1, respectively, and induced a higher secretion of proinflammatory cytokines interleukin-2 (IL-2) and interferon-gamma (IFNγ) in vitro. In addition, bioluminescence imaging of tumor xenografts of ACAT-1-inhibited targeting MSLN CAR-T cells in MSLN-positive PC mice in vivo showed significant tumor regression, which is consistent with the in vitro observations. Our findings demonstrate a novel immunotherapeutic strategy involving the transplantation of ACAT-1-inhibited targeting MSLN CAR-T cells and the feasibility of enhancing the antitumor potency of CAR-T through the novel strategy.

6.
Front Pharmacol ; 11: 598170, 2020.
Article in English | MEDLINE | ID: mdl-33390986

ABSTRACT

Background: Drugs can alter the gut microbiota structure, and gut microbiota dysbiosis in turn is correlated with drug side effects through the intestinal endotoxemia hypothesis. Whether antithyroid drugs (including methimazole and propylthiouracil) cause gut microbiota dysbiosis and whether the gut microbiota is correlated with antithyroid drugs induced liver injury is unknown. Methods: Initial Graves' disease patients were randomly divided into the methimazole group (n = 20) and the propylthiouracil group (n = 20) and were followed up every 2 weeks; 50 healthy controls were also included. The structure and function of gut microbiota were compared from the cross sectional and longitudinal levels. The correlation between the gut microbiota and clinical parameters was also determined. In addition, Sprague-Dawley rats were randomly allotted into six groups, including four drug groups, which received daily doses of methimazole (1.5 mg/kg/day; 2.5 mg/kg/day) or propylthiouracil (7.5 mg/kg/day; 12.5 mg/kg/day) by oral gavage, and two control groups received the vehicle. In addition to the indexes mentioned above, intestinal barrier-related indexes were also performed. Results: Cross sectional and longitudinal comparison results from both clinical trials and animal studies indicate that antithyroid drugs altered gut microbiota structure; and the liver function related indexes all increased which correlated with gut microbiota. In addition, lipopolysaccharide-related pathways and the lipopolysaccharide concentration in feces and serum all increased after antithyroid drugs administration. These results consistent with the destroyed intestinal barrier in animal study after antithyroid drugs administration. Conclusion: We verified that antithyroid drugs altered gut microbiota structure and that the gut microbiota may in turn be correlated with antithyroid drugs-induced liver injury through the intestinal endotoxemia hypothesis.

8.
Int J Biochem Cell Biol ; 108: 98-110, 2019 03.
Article in English | MEDLINE | ID: mdl-30682488

ABSTRACT

Cholangiocarcinoma (CCA) is one of the most common fatal carcinomas and is well known to be lack of effective treatment. Thus, novel therapeutic strategies are greatly needed. Evodiamine, a quinozole alkaloid isolated from evodia rutaecarpa Bentham, has been demonstrated to exhibit anti-tumor effects on many cancer cells. However, little is known in terms of the effects on cholangiocarcinoma. In this study, we studied whether this traditional Chinese Medicine could serve as new potential therapeutic drugs to treat CCA. We discovered that evodiamine inhibited CCA cell proliferation and induced apoptosis. Moreover, evodiamine inhibited CCA cell migration and invasion. Mechanistically, our studies demonstrated that evodiamine inhibited the activation of IL-6 -induced STAT3 signaling activation, and the inhibitory effect was likely due to the upregulation of phosphatase shatterproof 2 (SHP-2), a negative feedback regulator of IL-6/STAT3. Blockage of SHP-2 through small interference RNA (siRNA) abolished the evodiamine -induced IL-6/STAT3 signaling inhibition. Moreover, in vivo experiment showed evodiamine inhibited the tumor growth of nude mice bearing TFK-1 xenografts. In summary, our results implied evodiamine as a promising anti-cancer agent in the treatment of CCA, and the mechanism is likely due to the inhibition of IL-6/STAT3 signaling with upregulating the expression levels of SHP-2.


Subject(s)
Antineoplastic Agents/pharmacology , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/biosynthesis , Quinazolines/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Induction/drug effects , Humans , Interleukin-6/metabolism , Mice , Neoplasm Invasiveness , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
9.
Environ Microbiol ; 21(2): 772-783, 2019 02.
Article in English | MEDLINE | ID: mdl-30548192

ABSTRACT

Colorectal cancer (CRC) is a common disease worldwide that is strongly associated with the gut microbiota. However, little is known regarding the gut microbiota after surgical treatment. 16S rRNA gene sequencing was used to evaluate differences in gut microbiota among colorectal adenoma patients, CRC patients, CRC postoperative patients and healthy controls by comparing gut microbiota diversity, overall composition and taxonomic signature abundance. The gut microbiota of CRC patients, adenoma patients and healthy controls developed in accordance with the adenoma-carcinoma sequence, with impressive shifts in the gut microbiota before or during the development of CRC. The gut microbiota of postoperative patients and CRC patients differed significantly. Subdividing CRC postoperative patients according to the presence or absence of newly developed adenoma which based on the colonoscopy findings revealed that the gut microbiota of newly developed adenoma patients differed significantly from that of clean intestine patients and was more similar to the gut microbiota of carcinoma patients than to the gut microbiota of healthy controls. The alterations of the gut microbiota between the two groups of postoperative patients corresponded to CRC prognosis. More importantly, we used the different gut microbiota as biomarkers to distinguish postoperative patients with or without newly developed adenoma, achieving an AUC value of 0.72. These insights on the changes in the gut microbiota of CRC patients after surgical treatment may allow the use of the microbiota as non-invasive biomarkers for the diagnosis of newly developed adenomas and to help prevent cancer recurrence in postoperative patients.


Subject(s)
Bacteria/isolation & purification , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/surgery , Gastrointestinal Microbiome , Adult , Aged , Aged, 80 and over , Bacteria/classification , Bacteria/genetics , Female , Humans , Intestines/microbiology , Male , Middle Aged , Young Adult
10.
Int J Cancer ; 144(11): 2728-2745, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30565661

ABSTRACT

The aim of our study was to investigate the relationship among the gut microbiota community, metabolite profiles and thyroid carcinoma (TC). First, 30 TC patients and 35 healthy controls (HCs) fecal samples were applied to characterize the gut microbial community using 16S rRNA gene sequencing. Differential microbiota compositions were observed, with significant enrichment of 19 and depletion of 8 genera in TC samples compared to those in HCs (Q value <0.05), and some genera were correlated with various clinical parameters, such as lipoprotein A and apolipoprotein B. Furthermore, 6 different genera distinguished TC patients from HCs with the AUC of 0.94. The PICRUSt analysis showed 12 remarkably different metabolic pathways (Q value <0.05). Subsequently, we systematically analyzed the gut microbiota and metabolites in the same TC patients (n = 15) and HCs (n = 15). The characteristics of the gut microbiota community were mostly consistent with the above results (30 TC patients and 35 HCs), and liquid chromatography mass spectrometry analysis was performed to characterize the metabolite profiles. In total, 21 different genera (Q value <0.05) and 72 significantly changed metabolites (VIP > 1.0 and p < 0.05) were observed and correlated to each other. Eight metabolites combined with 5 genera were more effective in distinguishing TC patients from HCs (AUC = 0.97). In conclusion, our study presents a comprehensive landscape of the gut microbiota and metabolites in TC patients, and provides a research direction of the mechanism of interaction between gut microbiota alteration and TC pathogenesis.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Thyroid Cancer, Papillary/microbiology , Thyroid Neoplasms/microbiology , Adult , Case-Control Studies , Cohort Studies , DNA, Bacterial/isolation & purification , Female , Healthy Volunteers , Humans , Intestinal Mucosa/microbiology , Male , Metabolomics , Middle Aged , RNA, Ribosomal, 16S/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology
11.
Emerg Microbes Infect ; 7(1): 197, 2018 Nov 30.
Article in English | MEDLINE | ID: mdl-30498207

ABSTRACT

Splenectomy carries a long-term risk of postoperative infection, and the chronic, low-grade inflammation associated with endotoxemia may be related to the gut microbiota. In this study, to increase our understanding of the potential cause of the high rate of infection in postsplenectomy patients, we evaluated the differences in the gut microbiota and plasma lipopolysaccharide level of patients after splenectomy relative to those of healthy controls. Thirty-two patients having undergone splenectomy and 42 healthy individuals were enrolled into the splenectomy (SP) and healthy control (HC) groups, respectively. The SP group was subdivided into three subgroups according to the length of their postoperative time. Fecal samples were used for gut microbiota analysis via 16s rRNA gene sequencing, blood examinations and plasma lipopolysaccharide measurements were also taken. Significant differences were observed in gut microbiota composition with regard to the relative bacterial abundances of 2 phyla, 7 families, and 15 genera. The lipopolysaccharide level was significantly higher in the SP group than in the HC group and were negatively associated with five bacterial families with low abundance in the SP group. The degree of the microbiota alteration increased with the length of the postoperative time. The PICRUSt analysis showed that the relative abundances of lipopolysaccharide biosynthesis proteins and lipopolysaccharide biosynthesis pathways were higher in the SP group and were positively associated with the plasma lipopolysaccharide level. Significant alterations were observed in the gut microbiota of the splenectomized patients and were associated with plasma lipopolysaccharide level. Further studies are needed to verify whether such alterations after splenectomy are related to an increased risk of complications.


Subject(s)
Bacteria/classification , Endotoxemia/etiology , Gastrointestinal Microbiome , Splenectomy/adverse effects , Acute-Phase Proteins/genetics , Adult , Bacteria/isolation & purification , Biosynthetic Pathways , Carrier Proteins/genetics , Case-Control Studies , Feces/microbiology , Female , Humans , Lipopolysaccharides/blood , Male , Membrane Glycoproteins/genetics , Middle Aged , RNA, Ribosomal, 16S/genetics
12.
Front Microbiol ; 9: 963, 2018.
Article in English | MEDLINE | ID: mdl-29867867

ABSTRACT

Dysbiosis of gut microbiota and metabolome is a frequently encountered condition in liver cirrhosis (LC) patients. The severity of liver dysfunction was found to be correlated with the degree of microbial dysbiosis. Several clinical studies have indicated liver function improvement after therapeutic splenectomy for LC-induced hypersplenism. We sought to determine whether such post-splenectomy outcome is pertinent to modulation of the abnormal gut microenvironment in LC patients. A cross-sectional study including 12 LC patients and 16 healthy volunteers was first conducted, then a before-after study in the cohort of patients was carried out before and 6 months after splenectomy. Fecal samples were collected in hospital. Temporal bacterial (n = 40) and metabolomics (n = 30) profiling was performed using 16s rRNA gene sequencing and ultra performance liquid chromatography/mass spectrometer (UPLC/MS), respectively. Our results revealed that microbial composition in patients was clearly different from that in healthy controls (HCs), evidenced by considerable taxonomic variation. Along with improved liver function (Child-Pugh score), the patients also displayed similar gut microbiota profile and predicted metagenome function to that of HCs after splenectomy. Enterobacteriaceae and Streptococcaceae, two LC-enriched families showing positive relation with Child-Pugh score, exhibited significantly decreased abundance after splenectomy. At the genus level, 11 genera were differentially abundant between patients and HCs, but 9 genera of them restituted to normal levels by certain degree after splenectomy. PICRUSt analysis showed that the relative abundance of 17 KEGG pathways was partially restored after splenectomy. Four of them were amino acid-related pathways: lysine degradation, tryptophan degradation, amino acid metabolism, and protein digestion and absorption. These findings were supported by metabonomics results which showed that relative abundance of amino acid and corresponding catabolites changed toward normal. In addition to the variations in the relative abundances of bacteria and metabolites, the correlation between them also altered in patients after splenectomy. Dysbiosis in gut microbiome and related metabolism of LC patients was partially corrected after splenectomy. Whether the improved gut microenvironment could prevent LC-related complications and delay the progress of LC is a propitious objective for future study. TRIAL REGISTRATION: ChiCTR-OOB-15007409. Registered November 15, 2015.

13.
Front Physiol ; 9: 470, 2018.
Article in English | MEDLINE | ID: mdl-29780327

ABSTRACT

Disturbance of the gut microbiota is common in liver cirrhosis (LC) patients, the underlying mechanisms of which are yet to be unfolded. This study aims to explore the relationship between small bowel transit (SBT) and gut microbiota in LC patients. Cross-sectional design was applied with 36 LC patients and 20 healthy controls (HCs). The gut microbiota was characterized by 16S rRNA gene sequencing. The Firmicutes/Bacteroidetes (F/B) ratio and the Microbial Dysbiosis index (MDI) were used to evaluate the severity of microbiota dysbiosis. The scintigraphy method was performed in patients to describe the objective values of SBT. Patients were then subdivided according to the Child-Pugh score (threshold = 5) or SBT value (threshold = 0.6) for microbiota analysis. LC patients were characterized by an altered gut microbiota; F/B ratios and MDI were higher than HC in both Child_5 (14.00 ± 14.69 vs. 2.86 ± 0.99, p < 0.01; 0.49 ± 0.80 vs. -0.47 ± 0.69, p < 0.01) and Child_5+ (15.81 ± 15.11 vs. 2.86±0.99, p < 0.01; 1.11 ± 1.05 vs. -0.47 ± 0.69, p < 0.01) sub-groups in patients. Difference in the gut microbiota between Child_ 5 and Child_5+ patients was inappreciable, but the SBT was relatively slower in Child_5+ patients (43 ± 26% vs. 80 ± 15%, p < 0.05). Compared with the Child-Pugh score indicators, SBT showed stronger associations with bacterial genera. A clear difference in the gut microbiota was observed between SBT_0.6- and SBT_0.6+ patients [Pr(>F) = 0.0068, pMANOVA], with higher F/B ratios and MDI in SBT_0.6- patients (19.71 ± 16.62 vs. 7.33 ± 6.65, p < 0.01; 1.02 ± 0.97 vs. 0.20 ± 0.58, p < 0.01). Similar results were observed between the SBT_0.6- and SBT_0.6+ sub-groups of patients with normal liver function and a Child-Pugh score of 5. SBT was negatively correlated with both the F/B ratio and MDI (r = -0.34, p < 0.05; r = -0.38, p < 0.05). Interestingly, an increased capacity for the inferred pathway "bacterial invasion of epithelial cells" in patients, was highly negatively correlated with SBT (r = -0.57, p < 0.01). The severity of microbiota dysbiosis in LC patients depends on SBT rather than Child-Pugh score. SBT per se might be significantly related to the gut microbiota abnormalities observed in patients with LC.

14.
Cell Physiol Biochem ; 47(2): 641-653, 2018.
Article in English | MEDLINE | ID: mdl-29794468

ABSTRACT

BACKGROUND/AIMS: Cholangiocarcinoma (CCA) is a malignant tumor that is resistant to chemotherapy, so new therapeutic agents are needed. Allicin which is rapidly converted from allin by allinase, is one of the most biologically active compounds in freshly crushed garlic and has been shown to have strong anti-tumor effects. Our aim was to explore the molecular mechanism by which allicin affects the cell proliferation and invasion of CCA. METHODS: Cell viability and apoptosis were measured using the CCK-8 assay, colony formation assay, and flow cytometry. Cell migration and invasion were evaluated by wound healing and Transwell assays, respectively. The expression of several proteins involved in cell apoptosis and invasion were assessed by Western blot. The activation of STAT3 signaling was detected by Western blot and immunofluorescence staining. The involvement of SHP-1 was determined using small interfering RNA (siRNA). Moreover, a nude mouse model of human CCA was established to assess the anti-tumor effects of allicin in vivo. RESULTS: Allicin significantly suppressed CCA cell proliferation by activating the caspase cascade, inducing apoptosis, and reducing the expression of proteins downstream of STAT3, such as B-cell lymphoma 2 (Bcl-2), while upregulating Bcl-2-associated X (Bax) protein. In addition, allicin inhibited the migration, invasion, and epithelial-mesenchymal transition (EMT) of CCA cells. Moreover, the protein expression of MMP-2 and MMP-9 was significantly downregulated in CCA cells treated with allicin compared with CCA cells treated with control. Mechanistic investigations indicated that allicin upregulated SHP-1 expression in CCA, and pervanadate treatment reversed the allicin-induced downregulation of STAT3. Moreover, suppression of SHP-1 by siRNA overturned the effect of allicin on the induction of SHP-1 and inhibition of STAT3 activation. Additionally, treatment with allicin attenuated tumor growth in the nude mouse model of CCA. CONCLUSIONS: Our findings suggest that allicin suppresses cell proliferation and invasion via STAT3 signaling and may be a potential therapeutic agent for CCA.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sulfinic Acids/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Disulfides , Epithelial-Mesenchymal Transition/drug effects , Humans , Matrix Metalloproteinases/metabolism , Mice , Mice, Nude , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/genetics , Sulfinic Acids/chemistry , Sulfinic Acids/metabolism , Sulfinic Acids/therapeutic use , Transplantation, Heterologous
15.
Thyroid ; 28(2): 175-186, 2018 02.
Article in English | MEDLINE | ID: mdl-29320965

ABSTRACT

BACKGROUND: Hashimoto's thyroiditis (HT) is an organ-specific autoimmune disease in which both genetic predisposition and environmental factors serve as disease triggers. Many studies have indicated that alterations in the gut microbiota are important environmental factors in the development of inflammatory and autoimmune diseases. A comparative analysis was systematically performed of the gut microbiota in HT patients and healthy controls. METHODS: First, a cross-sectional study of 28 HT patients and 16 matched healthy controls was conducted. Fecal samples were collected, and microbiota were analyzed using 16S ribosomal RNA gene sequencing. Second, an independent cohort of 22 HT patients and 11 healthy controls was used to evaluate the diagnostic potential of the selected biomarkers. RESULTS: Similar levels of bacterial richness and diversity were found in the gut microbiota of HT patients and healthy controls (p = 0.11). A detailed fecal microbiota Mann-Whitney U-test (Q value <0.05) revealed that the abundance levels of Blautia, Roseburia, Ruminococcus_torques_group, Romboutsia, Dorea, Fusicatenibacter, and Eubacterium_hallii_group genera were increased in HT patients, whereas the abundance levels of Fecalibacterium, Bacteroides, Prevotella_9, and Lachnoclostridium genera were decreased. A correlation matrix based on the Spearman correlation distance confirmed correlations among seven clinical parameters. Additionally, the linear discriminant analysis effect size method showed significant differences in 27 genera between the two groups that were strongly correlated with clinical parameters. The linear discriminant analysis value was used to select the first 10 species from the 27 different genera as biomarkers, achieving area under the curve values of 0.91 and 0.88 for exploration and validation data, respectively. CONCLUSIONS: Characterization of the gut microbiota in HT patients confirmed that HT patients have altered gut microbiota and that gut microbiota are correlated with clinical parameters, suggesting that microbiome composition data could be used for disease diagnosis. Further investigation is required to understand better the role of the gut microbiota in the pathogenesis of HT.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome/physiology , Hashimoto Disease/microbiology , Adult , Cross-Sectional Studies , Female , Hashimoto Disease/blood , Humans , Male , Middle Aged , Thyroid Hormones/blood
16.
J Immunother ; 41(2): 45-52, 2018.
Article in English | MEDLINE | ID: mdl-29252915

ABSTRACT

Chimeric antigen receptor-modified T cell (CART) therapy has been demonstrated to have significant effect on hematologic tumor in patients. However, many persistent obstacles and challenges still limit the application. It is known that CD8 T cells are a key component of antitumor immunity. An avasimibe-induced inhibition of cholesterol esterification has been shown to improve the antitumor response of CD8 T cells in mice. In this study, using human CD19-directed CART cells as effector cells and CD19-overexpressing K562 cells as target cells, we detected whether cholesterol acyltransferase inhibition by avasimibe can enhance the antitumor effect of human CART cells. After avasimibe treatment, the infection rate was dropped by up to 50% (P<0.05). The cytotoxic effect of CART cells was significantly increased than the control group in a dose-dependent manner. Moreover, the level of secreted interferon-γ increased in almost half of the cases (P<0.05); the ratio of CD8CD4 T cells was increased among the total T cells and the CART cells in some of cases (P<0.05). Our study suggests that inhibition of cholesterol acyltransferase can promote the antitumor effect of CART cells, and provides a new option for a combination therapy by regulating T-cell metabolism to enhance antitumor effects.


Subject(s)
Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , Sterol O-Acyltransferase/antagonists & inhibitors , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Animals , Antigens, CD19/immunology , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Humans , Immunotherapy, Adoptive , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...