Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Front Oncol ; 13: 1257404, 2023.
Article in English | MEDLINE | ID: mdl-37588092

ABSTRACT

[This corrects the article DOI: 10.3389/fonc.2022.993243.].

4.
Transl Res ; 259: 1-12, 2023 09.
Article in English | MEDLINE | ID: mdl-36977441

ABSTRACT

Chimeric antigen receptor T cell (CAR-T) therapy has limited efficacy for treating glioma because of the infiltrative nature of the blood-brain barrier (BBB) and T cell exhaustion. Conjugation with rabies virus glycoprotein (RVG) 29 enhances the brain-related efficacy of various agents. Here we assess whether RVG enhances the ability of CAR-T cells to cross the BBB and improves their immunotherapy. We generated 70R CAR-T cells (anti-CD70 CAR-T modified with RVG29) and validated their tumor-killing efficacy in vitro and in vivo. We validated their effects on tumor regression in a human glioma mouse orthotopic xenograft model as well as in patient-derived orthotopic xenograft (PDOX) models. The signaling pathways activated in 70R CAR-T cells were revealed by RNA sequencing. The 70R CAR-T cells we generated showed effective antitumor function against CD70+ glioma cells both in vitro and in vivo. 70R CAR-T cells were better able to cross the BBB into the brain than CD70 CAR-T cells under the same treatment conditions. Moreover, 70R CAR-T cells significantly promote the regression of glioma xenografts and improve the physical characteristics of mice without causing overt adverse effects. RVG modification enables CAR-T cells to cross the BBB, and stimulation with glioma cells induces 70R CAR-T cells to expand in a resting state. The modification of RVG29 has a positive impact on CAR-T therapy for brain tumors and may have potential in CAR-T therapy for glioma.


Subject(s)
Glioma , Rabies virus , Receptors, Chimeric Antigen , Humans , Animals , Mice , Glioma/therapy , Glioma/metabolism , Glycoproteins , Immunotherapy, Adoptive , Xenograft Model Antitumor Assays , Cell Line, Tumor
5.
Front Oncol ; 12: 993243, 2022.
Article in English | MEDLINE | ID: mdl-36439421

ABSTRACT

The expression of O-GlcNAc transferase (OGT) and its catalytic product, O-GlcNAcylation (O-GlcNAc), are elevated in many types of cancers, including prostate cancer (PC). Inhibition of OGT serves as a potential strategy for PC treatment alone or combinational therapy. PC is the second common cancer type in male worldwide, for which chemotherapy is still the first-line treatment. However, the function of inhibition of OGT on chemotherapeutic response in PC cells is still unknown. In this study, we show that inhibition of OGT by genetic knockdown using shRNA or by chemical inhibition using OGT inhibitors sensitize PC cells to docetaxel, which is the most common chemotherapeutic agent in PC chemotherapy. Furthermore, we identified that microRNA-140 (miR-140) directly binds to OGT mRNA 3' untranslated region and inhibits OGT expression. Moreover, docetaxel treatment stimulates miR-140 expression, whereas represses OGT expression in PC cells. Overexpression of miR-140 enhanced the drug sensitivity of PC cells to docetaxel, which could be reversed by overexpression of OGT. Overall, this study demonstrates miR-140/OGT axis as therapeutic target in PC treatment and provides a promising adjuvant therapeutic strategy for PC therapy.

6.
Elife ; 112022 05 03.
Article in English | MEDLINE | ID: mdl-35502895

ABSTRACT

Methyltransferase-like 3 (METTL3) and N6-methyladenosine (m6A) are involved in many types of biological and pathological processes, including DNA repair. However, the function and mechanism of METTL3 in DNA repair and chemotherapeutic response remain largely unknown. In present study, we identified that METTL3 participates in the regulation of homologous recombination repair (HR), which further influences chemotherapeutic response in both MCF-7 and MDA-MB-231 breast cancer (BC) cells. Knockdown of METTL3 sensitized these BC cells to Adriamycin (ADR; also named as doxorubicin) treatment and increased accumulation of DNA damage. Mechanically, we demonstrated that inhibition of METTL3 impaired HR efficiency and increased ADR-induced DNA damage by regulating m6A modification of EGF/RAD51 axis. METTL3 promoted EGF expression through m6A modification, which further upregulated RAD51 expression, resulting in enhanced HR activity. We further demonstrated that the m6A 'reader,' YTHDC1, bound to the m6A modified EGF transcript and promoted EGF synthesis, which enhanced HR and cell survival during ADR treatment in BC. Our findings reveal a pivotal mechanism of METTL3-mediated HR and chemotherapeutic drug response, which may contribute to cancer therapy.


Subject(s)
Breast Neoplasms , Epidermal Growth Factor , Methyltransferases , Rad51 Recombinase , Recombinational DNA Repair , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Female , Humans , Methyltransferases/genetics , Methyltransferases/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
7.
J Hematol Oncol ; 14(1): 152, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556152

ABSTRACT

Chimeric antigen receptor T-cell (CAR-T) therapy has shown tremendous success in eradicating hematologic malignancies. However, this success has not yet been extrapolated to solid tumors due to the limited infiltration and persistence of CAR-T cells in the tumor microenvironment (TME). In this study, we screened a novel anti-CD70 scFv and generated CD70 CAR-T cells that showed effective antitumor functions against CD70+ renal carcinoma cells (RCCs) both in vitro and in vivo. We further evaluated the effect and explored the molecular mechanism of a PARP inhibitor (PARPi) in CAR-T cell immunotherapy by administering the PARPi to mouse xenografts model derived from human RCC cells. Treatment with the PARPi promoted CAR-T cell infiltration by stimulating a chemokine milieu that promoted CAR-T cell recruitment and the modulation of immunosuppression in the TME. Moreover, our data demonstrate that PARPi modulates the TME by activating the cGAS-STING pathway, thereby altering the balance of immunostimulatory signaling and enabling low-dose CAR-T cell treatment to induce effective tumor regression. These data demonstrate the application of CD70 CAR-T cell therapeutic strategies for RCC and the cross-talk between targeting DNA damage responses and antitumor CAR-T cell therapy. These findings provide insight into the mechanisms of PARPis in CAR-T cell therapy for RCC and suggest a promising adjuvant therapeutic strategy for CAR-T cell therapy in solid tumors.


Subject(s)
CD27 Ligand/antagonists & inhibitors , Carcinoma, Renal Cell/therapy , Immunotherapy, Adoptive/methods , Kidney Neoplasms/therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Single-Chain Antibodies/therapeutic use , Animals , CD27 Ligand/immunology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/immunology , Cell Line, Tumor , DNA Damage/drug effects , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Membrane Proteins/immunology , Mice , Nucleotidyltransferases/immunology , Signal Transduction
8.
Cell Death Dis ; 12(6): 503, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34006852

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 (APE1) plays a critical role in the base excision repair (BER) pathway, which is responsible for the excision of apurinic sites (AP sites). In non-small cell lung cancer (NSCLC), APE1 is highly expressed and associated with poor patient prognosis. The suppression of APE1 could lead to the accumulation of unrepaired DNA damage in cells. Therefore, APE1 is viewed as an important marker of malignant tumors and could serve as a potent target for the development of antitumor drugs. In this study, we performed a high-throughput virtual screening of a small-molecule library using the three-dimensional structure of APE1 protein. Using the AP site cleavage assay and a cell survival assay, we identified a small molecular compound, NO.0449-0145, to act as an APE1 inhibitor. Treatment with NO.0449-0145 induced DNA damage, apoptosis, pyroptosis, and necroptosis in the NSCLC cell lines A549 and NCI-H460. This inhibitor was also able to impede cancer progression in an NCI-H460 mouse model. Moreover, NO.0449-0145 overcame both cisplatin- and erlotinib-resistance in NSCLC cell lines. These findings underscore the importance of APE1 as a therapeutic target in NSCLC and offer a paradigm for the development of small-molecule drugs that target key DNA repair proteins for the treatment of NSCLC and other cancers.


Subject(s)
Apoptosis/immunology , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Necroptosis/immunology , Pyroptosis/immunology , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude
9.
Oncogene ; 39(33): 5507-5519, 2020 08.
Article in English | MEDLINE | ID: mdl-32641859

ABSTRACT

DNA polymerase ß (Pol ß) plays a critical role in DNA base excision repair (BER), which is involved in maintaining genomic stability and in the modulation of DNA demethylation. Numerous studies implicated deficiency of Pol ß in the genomic instability and dysregulation of genes expression, leading to affecting initiation of cancer. However, the role of Pol ß in cancer progression is still unclear. Here, we show that Pol ß depresses migratory and invasive capabilities of both breast and lung carcinomas, which were evident in human breast and lung cancer cells, as well as in mouse xenograft tumors. On the molecular basis, overexpression of Pol ß enhanced expression of CDH13, which show function on cell adhesion and migration. Knockdown of CDH13 restores the migratory, invasive capabilities and angiogenesis in tumor, which gets impaired by Pol ß. According to the function of BER on modulation of DNA demethylation, our studies on CDH13 expression and the DNA methylation levels of CDH13 promoter suggested that Pol ß promotes expression of CDH13 by augmenting DNA demethylation of CDH13 promoter. Our findings elucidated a novel possibility that Pol ß impair cancer cell metastasis during cancer progression and shed light on the role of Pol ß in cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Cadherins/metabolism , DNA Methylation , DNA Polymerase beta/metabolism , Lung Neoplasms/metabolism , A549 Cells , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cadherins/genetics , DNA Polymerase beta/genetics , Disease Models, Animal , Disease Progression , Female , Heterografts , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MCF-7 Cells , Mice , Mice, Nude , Mice, SCID , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...