Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
BMC Gastroenterol ; 24(1): 151, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698325

ABSTRACT

BACKGROUND: Acute pancreatitis (AP) is a prevalent exocrine inflammatory disorder of the pancreas characterized by pancreatic inflammation and injury to acinar cells. Vitamin B6 (VB6) is a vital nutrient that plays a significant role in preserving human health and has anti-inflammatory and anti-apoptotic effects. METHODS: This study aimed to explore the potential pancreatic protective effects of VB6 in mitigating pancreatic inflammation and apoptosis induced by taurocholate sodium (TLCS) in an AP model and to assess the underlying mechanism of action. AP was induced in Sprague‒Dawley (SD) rats through TLCS administration and lipopolysaccharide (LPS)-treated AR42J cells, followed by treatment with VB6. RESULTS: Various parameters associated with AP were assessed in both plasma and pancreatic tissues. VB6 has been shown to ameliorate the severity of AP through various mechanisms. It effectively reduces the levels of serum amylase, lipase, and inflammatory factors, thereby mitigating histological injury to the pancreas. Moreover, VB6 inhibited pancreatic apoptosis by downregulating bax expression and up-regulating Bcl2 expression in TLCS-treated rats. Additionally, VB6 suppressed the expression of caspase3. The anti-inflammatory and anti-apoptotic effects of VB6 observed in LPS-treated AR42J cells are consistent with those observed in a rat model of AP. CONCLUSIONS: These results suggest that VB6 exerts anti-inflammatory and anti-apoptotic effects through inhibition of the caspase3 signaling pathway and has a protective effect against AP.


Subject(s)
Apoptosis , Caspase 3 , Lipopolysaccharides , Pancreatitis , Rats, Sprague-Dawley , Signal Transduction , Taurocholic Acid , Vitamin B 6 , Animals , Pancreatitis/drug therapy , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis/chemically induced , Signal Transduction/drug effects , Apoptosis/drug effects , Caspase 3/metabolism , Rats , Vitamin B 6/pharmacology , Vitamin B 6/therapeutic use , Male , Amylases/blood , Pancreas/pathology , Pancreas/drug effects , Pancreas/metabolism , Disease Models, Animal , Anti-Inflammatory Agents/pharmacology , Acute Disease , bcl-2-Associated X Protein/metabolism , Lipase/metabolism , Lipase/blood , Proto-Oncogene Proteins c-bcl-2/metabolism
2.
Med Oncol ; 40(3): 90, 2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36737590

ABSTRACT

The purpose of this study was to investigate the effect of the interaction between tumor-associated fibroblasts (CAFs) and macrophage polarization on colorectal cancer (CRC) progression. Clinical tissue samples of CRC and health volunteers were collected to isolate normal fibroblasts (NFs) and CAFs. LoVo, HCT116, or THP-1 cells were co-cultured with NFs or CAFs. Immunofluorescence and western blot detected the expression of related markers. MTT assay measured cell viability and IC50. Cell proliferation and metastasis were detected through colony formation and transwell assays. CRC mice models were constructed by injection of HCT116 cells, with IHC assessing C-X-C Motif Chemokine Ligand 12 (CXCL12) expression. The proliferation, migration, invasion, and cisplatin (DDP) resistance of CRC cells were apparently increased after co-culture with CAFs. Compared to NFs, CAFs have a markedly higher ability to recruit macrophages and promote macrophages M2 polarization by secreting CXCL12. Further experiments affirmed that CXCL12 secreted by CAFs boosted proliferation, migration, invasion, and DDP resistance of CRC cells via induction of the M2 polarization of macrophages. In vivo experiments confirmed that CAFs promoted the progression of CRC and DDP resistance by affecting M2 polarization through CXCL12. CAFs recruit macrophages and secrete CXCL12 to induce M2 polarization of macrophages, thus mediating cell function and DDP resistance of CRC.


Subject(s)
Cisplatin , Colorectal Neoplasms , Animals , Mice , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Cisplatin/pharmacology , Colorectal Neoplasms/pathology , Macrophages/metabolism , Tumor Microenvironment , Fibroblasts/metabolism , Chemokine CXCL12/metabolism
3.
Anticancer Drugs ; 34(3): 422-430, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36730310

ABSTRACT

The purpose of this study was to investigate the effect of chemoresistant cancer-associated fibroblasts (R-CAFs) against cisplatin (DDP) on colorectal cancer (CRC) progression. First, clinical tissue samples of chemoresistant or chemosensitive CRC patients were collected to isolate R-CAFs or chemosensitive CAFs (S-CAFs), respectively. HT29 cells or HUVECs were co-cultured with R-CAFs by transwell device. Then the proliferation and apoptosis of HT29 cells were detected with Cell Counting Kit-8 (CCK-8) and flow cytometry. Transwell assay and tube formation assay was used to detect the migration and angiogenesis of HUVECs. In addition, a colorectal cancer transplantation model was established subcutaneously in nude mice by injecting stably transfected HT29 cells and exosomes from different CAF groups, and then the tumor volume and weight were measured and recorded. Hematoxylin and eosin staining, immunohistochemistry, and terminal deoxynucleotidyl transferase dUTP Nick-End Labeling (TUNEL) staining were performed to characterize the histopathological characteristics and apoptosis level of tumor tissues, respectively. S-CAFs and R-CAFs were isolated successfully. HT29 cell co-culture with R-CAFs significantly affected the proliferation and apoptosis of HT29 cells. Exosomes derived from R-CAFs (R-CAFs-Exo) were delivered to HT29 cells, which could induce viability, suppress apoptosis and accelerate the angiogenesis of CRC. In addition, VEGFA was highly expressed in R-CAFs-Exo, which might indicate that R-CAFs could transmit VEGFA through exosomes. Overexpressed VEGFA in R-CAFs apparently regulates the viability, apoptosis, DDP resistance, and angiogenesis of CRC. In-vivo experiments confirmed that R-CAFs-Exo promoted the progression of CRC and DDP resistance by delivering VEGFA . R-CAFs-derived exosomes promote the viability, apoptosis, DDP resistance, and angiogenesis of CRC by delivering VEGFA .


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Exosomes , MicroRNAs , Animals , Mice , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cell Proliferation , Cisplatin , Colorectal Neoplasms/pathology , Mice, Nude , Humans
4.
Int Immunopharmacol ; 114: 109450, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36446233

ABSTRACT

CD46, CD55 and CD59 are membrane-bound complement regulatory proteins (mCRPs) and highly expressed in many tumor tissues. Our analysis by RNA sequencing and qRT-PCR revealed that the expression of mCRPs was significantly elevated in cancer tissues of 15 patients with colon cancer. To further investigate the role of mCRPs in the development of colon cancer, we suppressed the expression of mCRPs by CD46-shRNA, CD55-shRNA and CD59-shRNA in colon cancer cell lines, SW620 and HT-29 cells. The results indicated that CD46-shRNA, CD55-shRNA and CD59-shRNA effectively reduced the expression of mCRPs, accompanied with the increased LDH release and the percentage of Annexin V + 7-AAD- early phase of apoptotic cells. The similar cytotoxic effects were also observed in the cells treated with CD46 neutralizing antibody (aCD46), associated with the increased C5b-9 deposition, cleaved caspase-3 and Bax expression in the treated cells. The cytotoxic effects by mCRPs knock-down were potentiated in the cells co-treated with doxorubicin (Dox). In addition, STAT3, STAT6, and p38 MAPK inhibitors, including C188-9, AS1517499 and SB203580 effectively reduced the expression of CD46 in the treated colon cells, associated with increased cell apoptosis and LDH release. Further study with mouse model revealed that mCRPs knockdown by mCRPs-shRNA significantly reduced colon cancer growth, associated with increased expression of Bax, cleaved caspase-3 and C5b-9 deposition, but reduced expression of Bcl-2, IL-6 and IL-1beta in tumor tissues of nude mice transplanted with SW620 cells. Thereby, mCRPs expression in human colon cancer cells were upregulated by STAT3/STAT6/p38 MAPK signaling and mCRPs knockdown reduced colon cancer growth in mice through inducing tumor cell apoptosis.


Subject(s)
Colonic Neoplasms , Complement Membrane Attack Complex , Humans , Animals , Mice , Caspase 3 , Mice, Nude , bcl-2-Associated X Protein , Complement Activation , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Complement System Proteins/metabolism , Colonic Neoplasms/drug therapy , CD55 Antigens/genetics , CD55 Antigens/metabolism , Immunologic Factors , RNA, Small Interfering/genetics
5.
Cell Death Dis ; 12(12): 1121, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34853296

ABSTRACT

Long noncoding RNAs (lncRNAs) are regarded as crucial regulators in tumor progression. Potassium two pore domain channel subfamily K member 15 and WISP2 antisense RNA 1 (KCNK15-AS1) has been confirmed to inhibit the migration and invasion of pancreatic cancer (PC) cells. However, its downstream mechanism and effect on other cellular functions in PC remain unknown. This study probed the function and potential mechanism of KCNK15-AS1 in PC cell growth. RT-qPCR and western blot were employed to measure gene expression in PC cells. ISH was applied to analyze KCNK15-AS1 expression in PC tissues. Functional assays were utilized to evaluate PC cell proliferation, apoptosis, migration and EMT. Mechanical experiments were adopted to detect gene interaction in PC cells. The obtained data indicated that KCNK15-AS1 was down-regulated in PC cells and tissues. Overexpressing KCNK15-AS1 hindered cell proliferation, migration and EMT while facilitated cell apoptosis in PC. Mechanically, alkylation repair homolog protein 5 (ALKBH5) was verified to induce m6A demethylation of KCNK15-AS1 to mediate KCNK15-AS1 up-regulation. KCNK15-AS1 combined with KCNK15 5'UTR to inhibit KCNK15 translation. Moreover, KCNK15-AS1 recruited MDM2 proto-oncogene (MDM2) to promote RE1 silencing transcription factor (REST) ubiquitination, thus transcriptionally upregulating phosphatase and tensin homolog (PTEN) to inactivate AKT pathway. In conclusion, our study first confirmed that KCNK15-AS1 hinders PC cell growth by regulating KCNK15 and PTEN, suggesting KCNK15-AS1 as a potential biomarker of PC.


Subject(s)
AlkB Homolog 5, RNA Demethylase/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Humans , Mice , Pancreatic Neoplasms/pathology , Signal Transduction , Transfection , Up-Regulation , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...