Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
2.
Drug Resist Updat ; 61: 100822, 2022 03.
Article in English | MEDLINE | ID: mdl-35257981

ABSTRACT

Cancer cell metabolism including aerobic glycolysis, amino acid and fatty acid metabolism, has been extensively studied. Metabolic reprogramming is a major hallmark of cancer, which promotes cancer cell proliferation, progression and metastasis, as well as provokes resistance to chemotherapeutic drugs. Several signal transduction pathways, such as BCR, MEK/ERK, Notch, NF-κB and PI3K/AKT/mTOR, regulate tumor metabolism, hence promoting tumor cell growth, proliferation and progression. Therefore, targeting metabolic enzymes, metabolites or their signal transduction pathways may constitute a promising therapeutic strategy to enhance cancer treatment efficacy. Diffuse large B-cell lymphoma (DLBCL) is the most aggressive form of non-Hodgkin lymphoma (NHL), and one-third of DLBCL patients suffer from relapsed/refractory disease after chemotherapy. The mechanisms underlying drug resistance are complex, including target gene mutations, metabolic reprogramming, aberrant signal transduction pathways, enhanced drug efflux via overexpression of multidrug efflux transporters like P-glycoprotein, upregulation of anti-apoptotic proteins, drug sequestration and enhanced DND repair. This review delineates the distinct metabolic reprogramming patterns and the association between metabolism and anticancer drug resistance in DLBCL as well as the emerging strategies to surmount chemoresistance in DLBCL.


Subject(s)
Drug Resistance, Neoplasm , Lymphoma, Large B-Cell, Diffuse , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/therapeutic use , Signal Transduction/genetics
3.
Open Med (Wars) ; 16(1): 464-471, 2021.
Article in English | MEDLINE | ID: mdl-33817324

ABSTRACT

Gestational diabetes mellitus (GDM) is a metabolic dysregulation closely related to both obesity and type 2 diabetes; however, the molecular mechanism underlying GDM is still unclear. The purpose of this study was to investigate the effects of microRNA-377 (miR-377-3p) and fibronectin type III domain containing 5 (FNDC5) in regulating the cell growth of trophoblasts under high glucose (HG) conditions during the development of GDM. Serum miR-377-3p was upregulated and positively correlated with fasting blood glucose level in GDM patients. miR-377-3p downregulation increased the cell vitality and suppressed the cell apoptosis of HG-treated HTR-8/SVneo and BeWo cells. Using TargetScan prediction, luciferase assay, and western blot, it was found that miR-377-3p could target FNDC5 and suppress its expression. However, FNDC5 downregulation abolished the effect of miR-377-3p inhibitor in HTR-8/SVneo cells. Together, miR-377 is a potential target for GDM biomarker, which promotes cell growth and suppresses cell apoptosis, partly through the upregulation of FNDC5.

4.
Front Immunol ; 11: 564099, 2020.
Article in English | MEDLINE | ID: mdl-33329526

ABSTRACT

Background: The administration of second- or third-generation anti-CD19 chimeric antigen receptor (CAR) T cells has remarkably improved the survival of patients with relapsed or refractory B cell malignancies. However, there are limited clinical results from fourth-generation CAR-T cell therapy, and the factors affecting response rate and survival have not been fully determined. Methods: Lymphoma patients with progression or relapse after intensive treatments, including hematopoietic stem cell transplantation, and life expectancy >2 months were enrolled in the study. Peripheral lymphocytes were collected through apheresis, and magnetically selected T cells were lentivirally transduced with a 4th-generation CAR featuring an anti-CD19 CAR and the iCasp9 suicide switch (4SCAR19). The patients received 4SCAR19 T cell infusion after approximately seven days of expansion and a conditioning regimen comprising cyclophosphamide/fludarabine. The efficacy, safety, and risk factors were evaluated. Results: A total of 21 patients with relapsed/refractory B cell non-Hodgkin lymphoma were enrolled and received 4SCAR19 T cell infusions at a median dose of 8.9×105 CAR-T cells/kg. The overall response rate was 67% [95% confidence interval (CI), 43 to 85], with 43% of patients achieving a complete response and 24% having a partial response. The overall and complete response rates were 58 and 33% in the diffuse large B-cell lymphoma (DLBCL) group and 78 and 56% in the non-DLBCL group, respectively. The median overall survival was 23.8 months (95% CI, not reached), with a median follow-up of 13.7 months. Factors affecting overall survival were International Prognostic Index (IPI), disease type, and remission status after CAR-T cell treatment. The most common adverse events of grade 3 or 4 during treatment were neutropenia (76%), leukopenia (71%), and thrombocytopenia (29%). The incidence of cytokine release syndrome (CRS) was 14%, and all cases were grade 1. One patient developed grade 3 neurotoxicity. No deaths were attributed to infusion of 4SCAR19 T cells, CRS, or neurotoxicity. Conclusions: In this study, patients with relapsed or refractory B cell non-Hodgkin's lymphoma who received 4SCAR19 T cell therapy had durable responses and few of adverse events. The IPI model is suitable for evaluating the prognosis of patients receiving CAR-T cell therapy. Trial registration: Chinese Clinical Trial Registry (http://www.chictr.org.cn): ChiCTR-OOC-16007779.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/therapy , Neoplasm Recurrence, Local/therapy , Receptors, Chimeric Antigen/immunology , Adult , Aged , Cytokine Release Syndrome/etiology , Female , Follow-Up Studies , Humans , Leukopenia/etiology , Male , Middle Aged , Neutropenia/etiology , Prognosis , Progression-Free Survival , Risk Factors , Survival Rate , Thrombocytopenia/etiology
5.
Surg Obes Relat Dis ; 14(11): 1686-1690, 2018 11.
Article in English | MEDLINE | ID: mdl-30220645

ABSTRACT

BACKGROUND: Few studies of single-anastomosis duodeno-ileal bypass with sleeve gastrectomy (SADI-S) as the revision surgery for laparoscopic adjustable gastric banding (LAGB) have been published. OBJECTIVES: To explore the efficacy and safety of SADI-S as the revision surgery for LAGB. SETTING: The research was completed by the University Hospital. METHODS: From November 2013 to November 2015, a total of 22 weight-regain patients who previously underwent LAGB received SADI-S as the revision surgery at the People's Liberation Army General Hospital. Preoperative clinical characteristics as well as the data at 1, 3, 6, 12, 18, and 24 months after operation were collected and analyzed. RESULTS: The operation time of SADI-S was 105 ± 12.2 minutes, and intraoperative blood loss was 27.3 ± 5.8 mL. The percentage of excess weight loss was 20.55 ± 9.10%, 40.1 ± 6.02%, 63.52 ± 10.43%, 70.72 ± 8.54%, 78.34 ± 9.25%, and 81.57 ± 11.12% at 1, 3, 6, 12, 18 and 24 months after surgery, respectively. The 2-year complete remission rate of type 2 diabetes was 17 of 18, and the partial remission rate was 1 of 18 after operation. The glycated hemoglobin was 8.7% ± 1.1%, 7.7% ± .9%, 6.2% ± .6%, 5.7% ± .5%, 5.5% ± .6%, 6.0% ± .9%, and 5.7% ± .8% preoperatively and at 1, 3, 6, 12, 18, and 24 months after the operation, respectively. One case presented incisional hernia and was repaired. There was no conversion to laparotomy. Vitamins and trace elements were administrated long term to these patients after the operation, and no patients experienced vitamin or trace element deficiencies. CONCLUSION: SADI-S is safe and effective as a revision surgery for patients who experienced weight regain after LAGB. However, multicenter randomized controlled studies with larger sample sizes are needed to explore the long-term efficacy and safety of SADI-S.


Subject(s)
Duodenum/surgery , Gastrectomy/methods , Gastric Bypass/methods , Ileum/surgery , Reoperation/methods , Adult , Female , Gastrectomy/adverse effects , Gastric Bypass/adverse effects , Humans , Male , Obesity, Morbid/surgery , Postoperative Complications , Reoperation/adverse effects , Retrospective Studies , Young Adult
6.
J Exp Clin Cancer Res ; 37(1): 12, 2018 Jan 22.
Article in English | MEDLINE | ID: mdl-29357910

ABSTRACT

BACKGROUND: Oncogenic roles of epidermal growth factor receptor pathway substrate no.8 (EPS8) have been widely reported in various tumors, making targeting of EPS8 an appealing prospect. Here, we describe the role of EPS8 in acute myeloid leukemia (AML) and consider the potential of EPS8 as an anti-AML target. Nuclear localization signal (NLS) residues of tumor-associated proteins are crucial for cell cycle progression, and specific inhibitors derived from the NLS have inhibitory effect on cancer cells. The NLS in EPS8 has potential as a specific anti-AML target. METHODS: Gene Expression Omnibus expression profiles of AML patients were used to test associations between EPS8 expression and AML patient outcome. The biological characteristics of AML cells after EPS8 knockdown were analyzed in vitro and in vivo. A specific peptide (CP-EPS8-NLS) derived from the NLS of EPS8 (amino acids 298-310) was synthesized, and the anti-AML effects of CP-EPS8-NLS were analyzed in cancer cells and in xenograft models. Mutated CP-EPS8-NLS and penetratin served as controls. RESULTS: We observed that elevated EPS8 expression in AML patients is associated with poor outcome. Knockdown of EPS8 significantly suppressed the survival of AML cells in vitro and in vivo. CP-EPS8-NLS interfered with EPS8-associated signaling and consequently exerted anti-AML activity. Importantly, CP-EPS8-NLS displayed anti-AML activity in various AML cell types, with diminished activity in PBMCs. CP-ESP8-NLS suppressed U937 cell proliferation, and injection of CP-EPS8-NLS exerted potent antitumor activity in the xenograft tumor models. A synergistic effect of CP-EPS8-NLS and chemotherapeutic agents was also observed in vitro and in vivo. Mechanistically, treatment of various AML cells with CP-EPS8-NLS downregulated the expression of EPS8 and its downstream pathways. CONCLUSIONS: The function of CP-EPS8-NLS is explained by the presence of a NLS in EPS8, which has been shown to induce nuclear translocation, consequently resulting in EPS8 overexpression. These results indicate that EPS8 is a potential target for AML treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Antineoplastic Agents/pharmacology , Cell-Penetrating Peptides/pharmacology , Nuclear Localization Signals/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cell-Penetrating Peptides/chemical synthesis , Disease Models, Animal , Drug Synergism , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Signal Transduction/drug effects , Transcriptome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Oncol Rep ; 39(1): 119-128, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29192326

ABSTRACT

Although the introduction of tyrosine kinase inhibitors greatly improved the survival of patients with chronic myeloid leukemia (CML), drug resistance remains a problem. Thus, mechanism-based novel therapeutic targets warrant exploration. Recently, epidermal growth factor receptor kinase substrate 8 (EPS8), which has been identified as an oncogene and plays an important role in a broad spectrum of solid tumours, was reported to be related to poor prognosis or chemoresistance in acute leukemia patients. However, its role in CML remains unclear. In the present study, using q-RT­PCR, we demonstrated that CML patients expressed a higher level of EPS8 mRNA in bone marrow mononuclear cells than healthy controls. Then, to determine the effect of EPS8 on the biological functions of CML cells, EPS8 expression was knocked down in the human CML cell line K562. Reduced proliferation, increased apoptosis, impaired adhesion and migration were observed in K562 cells after EPS8 silencing. Notably, attenuation of EPS8 increased chemosensitivity both in imatinib-sensitive K562 cells and in the imatinib-resistant murine BCR-ABL+ 32D-p210BCR/ABL-T315I cells. Mechanistically, knockdown of EPS8 downregulated p-BCR/ABL and its downstream AKT/mTOR signalling pathway. Finally, knockdown of EPS8 attenuated K562 cell proliferation in BALB/c nude mice. These data indicated that EPS8 regulated the proliferation, apoptosis and chemosensitivity in BCR-ABL positive cells via the BCR-ABL/PI3K/AKT/mTOR pathway. Targeting EPS8 alone or combined with a tyrosine kinase inhibitor may be a promising alternative therapeutic strategy.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Signal Transduction , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Daunorubicin/pharmacology , Female , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic/drug effects , Humans , Imatinib Mesylate/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Male , Mice , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...