Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cancer Immunol Immunother ; 73(9): 164, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38954022

ABSTRACT

T cell receptor-engineered T cells (TCR-Ts) therapy is promising for cancer immunotherapy. Most studies have focused on identifying tumor-specific T cell receptors (TCRs) through predicted tumor neoantigens. However, current algorithms for predicting tumor neoantigens are unreliable and many neoantigens are derived from non-coding regions. Thus, the technological platform for identifying tumor-specific TCRs using natural antigens expressed on tumor cells is urgently needed. In this study, tumor organoids-enriched tumor infiltrating lymphocytes (oeT) were obtained by repeatedly stimulation of autologous patient-derived organoids (PDO) in vitro. The oeT cells specifically responded to autologous tumor PDO by detecting CD137 expression and the secretion of IFN-γ using enzyme-linked immunospot assay. The measurement of oeT cell-mediated killing of three-dimensional organoids was conducted using a caspase3/7 flow cytometry assay kit. Subsequently, tumor-specific T cells were isolated based on CD137 expression and their TCRs were identified through single-cell RT-PCR analysis. The specificity cytotoxicity of TCRs were confirmed by transferring to primary peripheral blood T cells. The co-culture system proved highly effective in generating CD8+ tumor-specific oeT cells. These oeT cells effectively induced IFN-γ secretion and exhibited specificity in killing autologous tumor organoids, while not eliciting a cytotoxic response against normal organoids. The analysis conducted by TCRs revealed a significant expansion of T cells within a specific subset of TCRs. Subsequently, the TCRs were cloned and transferred to peripheral blood T cells generation engineered TCR-Ts, which adequately recognized and killed tumor cell in a patient-specific manner. The co-culture system provided an approach to generate tumor-specific TCRs from tumor-infiltrating lymphocytes of patients with colorectal cancer, and tumor-specific TCRs can potentially be used for personalized TCR-T therapy.


Subject(s)
Coculture Techniques , Lymphocytes, Tumor-Infiltrating , Organoids , Receptors, Antigen, T-Cell , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Organoids/immunology , Antigens, Neoplasm/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/pathology
2.
J Cell Physiol ; 235(3): 1903-1914, 2020 03.
Article in English | MEDLINE | ID: mdl-31332789

ABSTRACT

Transforming growth factor ß (TGF-ß) is part of the transforming growth factor ß superfamily which is involved in many physiological processes and closely related to the carcinogenesis. Here, we discuss the TGF-ß structure, function, and its canonical Smads signaling pathway. Importantly, TGF-ß has been proved that it plays both tumor suppressor as well as an activator role in tumor progression. In an early stage, TGF-ß inhibits cell proliferation and is involved in cell apoptosis. In an advanced tumor, TGF-ß signaling pathway induces tumor invasion and metastasis through promoting angiogenesis, epithelial-mesenchymal transition, and immune escape. Furthermore, we are centered on updated research results into the inhibitors as drugs which have been studied in preclinical or clinical trials in tumor carcinogenesis to prevent the TGF-ß synthesis and block its signaling pathways such as antibodies, antisense molecules, and small-molecule tyrosine kinase inhibitors. Thus, it is highlighting the crucial role of TGF-ß in tumor therapy and may provide opportunities for the new antitumor strategies in patients with cancer.


Subject(s)
Neoplasms/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Neoplasms/drug therapy , Signal Transduction/drug effects
3.
J Cell Physiol ; 234(8): 13182-13190, 2019 08.
Article in English | MEDLINE | ID: mdl-30536619

ABSTRACT

Prostate cancer (CaP) is the second most common cancer in men worldwide in 2012, and radiation therapy is one of the most common definitive treatment options for localized CaP. However, radioresistance is a major challenge for the current radiotherapy, accumulating evidences suggest microRNAs (miRNAs), as an important regulator in cellular ionizing radiation (IR) responses, are closely correlated with radiosensitivity in many cancers. Here, we identified microRNA-16-5p(miR-16-5p) is significantly upregulated in CaP LNCaP cells following IR and can enhance radiosensitivity through modulating Cyclin D1/E1-pRb-E2F1 pathway. To identify the expression profile of miRNAs in CaP cells exposed to IR, we performed human miRNA probe hybridization chip analysis and miR-16-5p was found to be significantly overexpressed in all treatment groups that irradiated with different doses of X-rays and heavy ions (12 C6+ ). Furthermore, overexpression of miR-16-5p suppressed cell proliferation, reduced cell viability, and induced cell cycle arrest at G0/G1 phase, resulting in enhanced radiosensitivity in LNCaP cells. Additionally, miR-16-5p specifically targeted the Cyclin D1/E1-3'-UTR in LNCaP cells and affected the expression of Cyclin D1/E1 in both mRNA and protein levels. Taken together, miR-16-5p enhanced radiosensitivity of CaP cells, the mechanism may be through modulating Cyclin D1/Cyclin E1/pRb/E2F1 pathway to cause cell cycle arrest at G0/G1 phase. These findings provided new insight into the correlation between miR-16-5p, cell cycle arrest, and radiosensitivity in CaP, revealed a previously unrecognized function of miR-16-5p-Cyclin D1/E1-pRb-E2F1 regulation in response to IR and may offer an alternative therapy to improve the efficiency of conventional radiotherapy.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Radiation Tolerance/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin E/genetics , Cyclin E/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Humans , Male , MicroRNAs/genetics , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Prostatic Neoplasms/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction/genetics
4.
J Cell Physiol ; 234(8): 13014-13020, 2019 08.
Article in English | MEDLINE | ID: mdl-30537094

ABSTRACT

Irradiation (IR) can be used to treat cancer by inducing complex and irreparable DNA damage in the cancer cells, which may lead to their apoptotic death. However, little is known about the molecular mechanism of this DNA damage. Here, the non-small-cell lung cancer cell line A549 was treated with either X-ray or carbon ion combined with bleomycin (BLM). The cell survival rate, frequency of double-strand breaks (DSBs), dynamic changes in γH2AX, and p53 binding protein 1 (53BP1), and protein expression of Ku70, Rad51, and XRCC1 were determined by the clone formation assay, agarose gel electrophoresis, immunofluorescence, and western blot analysis. The results showed that the most obvious complex DSBs occurred in the carbon IR + BLM group. The number of γH2AX and 53BP1 foci in the 0.5 hr X-ray IR + BLM group was the highest (p < 0.001) among all the groups. γH2AX foci were detected in the nucleus at 0.5, 1, 2, and 4 hr, but were distributed throughout the cell at 6 hr after IR in the carbon ion IR + BLM group. The expression of Ku70 increased and XRCC1 decreased at 2 and 6 hr after IR. Our data indicate that a DNA damage frequency of 13.4/Mbp is caused by clustered DNA damage and further show a correlation between γH2AX, 53BP1, and XRCC1 levels and the extent of DNA damage. The results of this study provide insights into DNA damage recognition and a rationale for the clinical use of radiotherapy.


Subject(s)
DNA Damage , DNA Repair/physiology , Histones/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , X-ray Repair Cross Complementing Protein 1/metabolism , A549 Cells , Carcinoma, Non-Small-Cell Lung , DNA/radiation effects , Humans , Lung Neoplasms
5.
Oncoimmunology ; 12(1): 2265703, 2023.
Article in English | MEDLINE | ID: mdl-37808405

ABSTRACT

Chimeric antigen receptor (CAR) T cell immunotherapy has demonstrated success in the treatment of hematological malignancies; however, its efficacy and applications in solid tumors remain limited. Immunosuppressive factors, particularly inhibitory checkpoint molecules, restrict CAR T cell activity inside solid tumors. The modulation of checkpoint pathways has emerged as a promising approach to promote anti-tumor responses in CAR T cells. Programmed cell death protein 1 (PD1) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) are two critical immune-checkpoint molecules that suppress anti-tumor activity in T cells. Simultaneous targeting of these two inhibitory molecules could be an efficient checkpoint modulation strategy. Here, we developed a PD1-TIGIT chimeric immune-checkpoint switch receptor (CISR) that enhances the efficacy of CAR T cell immunotherapy by reversing the inhibitory checkpoint signals of PD1/PDL1 and/or TIGIT/CD155. In addition to neutralizing PDL1 and CD155, this chimeric receptor is engineered with the transmembrane region and intracellular domain of CD28, thereby effectively enhancing T cell survival and tumor-targeting functions. Notably, under simultaneous stimulation of PDL1 and CD155, CISR-CAR T cells demonstrate superior performance in terms of cell survival, proliferation, cytokine release, and cytotoxicity in vitro, compared with conventional CAR T cells. Experiments utilizing both cell line- and patient-derived xenotransplantation tumor models showed that CISR-CAR T cells exhibit robust infiltration and anti-tumor efficiency in vivo. Our results highlight the potential for the CISR strategy to enhance T cell anti-tumor efficacy and provide an alternative approach for T cell-based immunotherapies.


Subject(s)
Hematologic Neoplasms , Neoplasms , Humans , T-Lymphocytes , Programmed Cell Death 1 Receptor , Neoplasms/therapy , Immunotherapy , Hematologic Neoplasms/metabolism , Receptors, Immunologic/metabolism
6.
Cell Mol Immunol ; 20(5): 475-488, 2023 05.
Article in English | MEDLINE | ID: mdl-36941318

ABSTRACT

The activation of NLRC4 is a major host response against intracellular bacteria infection. However, NLRC4 activation after a host senses diverse stimuli is difficult to understand. Here, we found that the lncRNA LNCGM1082 plays a critical role in the activation of NLRC4. LNCGM1082 in macrophages affects the maturation of interleukin (IL)-1ß and pyroptotic cell death only after exposure to an NLRC4 ligand. Similar to NLRC4-/- mice, LNCGM1082-/- mice were highly sensitive to Salmonella Typhimurium (S. T) infection. LNCGM1082 deficiency in mouse or human macrophages inhibited IL-1ß maturation and pyroptosis. Mechanistically, LNCGM1082 induced the binding of PKCδ with NLRC4 in both mice and humans. In contrast, NLRC4 did not bind PKCδ in LNCGM1082-/- macrophages. The activity of the lncRNA LNCGM1082 induced by S. T may be mediated through TLR5 in the macrophages of both mice and humans. In summary, our data indicate that TLR5-mediated LNCGM1082 activity can promote the binding of PKCδ with NLRC4 to activate NLRC4 and induce resistance to bacterial infection.


Subject(s)
RNA, Long Noncoding , Salmonella Infections , Animals , Humans , Mice , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Inflammasomes/metabolism , Mice, Inbred C57BL , Toll-Like Receptor 5/metabolism
7.
J Crohns Colitis ; 16(6): 963-977, 2022 Jul 14.
Article in English | MEDLINE | ID: mdl-34964882

ABSTRACT

BACKGROUND AND AIMS: Increased E. coli in the colon are related to the occurrence and development of multiple diseases. Chemokines are shown to possess potential antimicrobial activity, including against Gram-positive and -negative bacterial pathogens. We here investigated function[s] of chemokine CXCL9 expressed in the gut epithelial cells, and mechanism[s] of CXCL9 by which to kill E. coli. METHODS: We generated CXCL9fl/flpvillin-creT mice [pvillin-cre positive mice] and their control CXCL9fl/flpvillin-crewmice [pvillin-cre negative mice], and then employed a dextran sulphate sodium [DSS]-mediated colitis model to determine the sensitivity of CXCL9fl/flpvillin-creT mice. We analysed the composition of the gut microbiota by using 16S ribosomal RNA [V3-V4 variable region] sequencing and shotgun metagenomic analyses. We generated E. coli ΔFtsX [FtsX-depleted E. coli] and E. coli ΔaceE [aceE-depleted E. coli] by using a bacterium red recombining system to investigate the mechanism[s] of CXCL9 by which to kill E. coli. RESULTS: CXCL9 fl/flpvillin-creTmice were more sensitive to chemically induced colitis than their control littermates, CXCL9fl/flpvillin-crewmice. After DSS treatment, there were markedly increased gut E. coli [Escherichia-Shigella] in the colonic contents of CXCL9fl/flpvillin-creT mice as compared with control CXCL9fl/flpvillin-crew mice. The increased E. coli could promote colitis through NLRC4 and caspase 1/11-mediated IL-18, which was derived from gut epithelial cells. We finally demonstrated that CXCL9 expressed in gut epithelial cells could kill the overgrown E. coli. E. coli expressed Ftsx and PDHc subunits aceE. E.coliΔaceE but not E. coliΔFtsX were resistant to CXCL9-mediated killing. CONCLUSIONS: Gut epithelial cells-derived CXCL9 can kill the expanded E. coli through aceE, to remain gut homeostasis.


Subject(s)
Colitis , Escherichia coli , Animals , Chemokine CXCL9/adverse effects , Colitis/genetics , Colon/microbiology , Dextran Sulfate , Disease Models, Animal , Homeostasis , Mice , Mice, Inbred C57BL
8.
Gut Microbes ; 14(1): 2139978, 2022.
Article in English | MEDLINE | ID: mdl-36519446

ABSTRACT

Antimicrobial proteins possess a broad spectrum of bactericidal activity and play an important role in shaping the composition of gut microbiota, which is related to multiple diseases such as metabolic syndrome. However, it is incompletely known for the regulation of defensin expression in the gut Paneth cells. Here, we found that FABP4 in the Paneth cells of gut epithelial cells and organoids can downregulate the expression of defensins. FABP4fl/flpvillinCreT mice were highly resistance to Salmonella Typhimurium (S.T) infection and had increased bactericidal ability to pathogens. The FABP4-mediated downregulation of defensins is through degrading PPARγ after K48 ubiquitination. We also demonstrate that high-fat diet (HFD)-mediated downregulation of defensins is through inducing a robust FABP4 in Paneth cells. Firmicutes/Bacteroidetes (F/B) ratio in FABP4fl/flpvillinCreT mice is lower than control mice, which is opposite to that in mice fed HFD, indicating that FABP4 in the Paneth cells could reprogram gut microbiota. Interestingly, FABP4-mediated downregulation of defensins in Paneth cells not only happens in mice but also in human. A better understanding of the regulation of defensins, especially HFD-mediated downregulation of defensin in Paneth cells will provide insights into factor(s) underlying modern diseases.Abbreviations: FABP4: Fatty acid binding protein 4; S. T: Salmonella Typhimurium; HFD: High-fat diet; Defa: α-defensin; 930 HD5: Human α-defensin 5; HD6: Human α-defensin 6; F/B: Firmicutes/Bacteroidetes; SFB: Segmental filamentous bacteria; AMPs: Antimicrobial peptides; PPARγ: Peroxisome proliferator-activated receptor γ; P-PPAR: Phosphorylated PPAR; Dhx15: DEAD-box helicase 15; 935 EGF: Epidermal growth factor; ENR: Noggin and R-spondin 1; CFU: Colony forming unit; Lyz1: Lysozyme 1; Saa1: Serum amyoid A 1; Pla2g2a: Phospholipase A2, group IIA; MMP-7: Matrix metalloproteinase; AU-PAGE: Acid-urea polyacrylamide gel electrophoresis; PA: Palmitic 940 acid; GPR40: G-protein-coupled receptor; GF: Germ-free; EGF: Epidermal growth factor; LP: Lamina propria; KO: Knock out; WT: Wild-type.


Subject(s)
Anti-Infective Agents , Fatty Acid-Binding Proteins , Gastrointestinal Microbiome , Animals , Humans , Mice , Anti-Infective Agents/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Paneth Cells/metabolism , RNA Helicases/metabolism
9.
Microbiome ; 10(1): 13, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35074011

ABSTRACT

BACKGROUND: IL-35-producing Bregs and Treg cells critically regulate chronic illnesses worldwide via mechanisms related to disrupting the gut microbiota composition. However, whether the gut microbiota regulates these IL-35+ cells remains elusive. We herein investigated the regulatory effects of the gut microbiota on IL-35+ cells by using genetically modified mouse models of obesity. RESULTS: We first found that gut Reg4 promoted resistance to high-fat diet-induced obesity. Using 16S rRNA sequencing combined with LC-MS (liquid chromatography-mass spectrometry)/MS, we demonstrated that gut Reg4 associated with bacteria such as Lactobacillus promoted the generation of IL-35+ B cells through 3-idoleacetic acid (IAA) in the presence of LPS. HuREG4IECtg mice fed a high-fat diet exhibited marked IL-35+ cell accumulation in not only their adipose tissues but also their colons, whereas decreased IL-35+ cell accumulation was observed in the adipose and colon tissues of Reg4 knockout (KO) mice. We also found that Reg4 mediated HFD-induced obesity resistance via IL-35. Lower levels of IAA were also detected in the peripheral blood of individuals with obesity compared with nonobese subjects. Mechanistically, IAA together with LPS mediated IL-35+ B cells through PXR and TLR4. KO of PXR or TLR4 impaired the generation of IL-35+ B cells. CONCLUSION: Together, IAA and LPS induce the generation of IL-35+ B cells through PXR and TLR4. Video Abstract.


Subject(s)
B-Lymphocytes , Gastrointestinal Microbiome , Interleukins , Lipopolysaccharides , Animals , Diet, High-Fat , Gastrointestinal Microbiome/physiology , Mice , Mice, Inbred C57BL , Obesity , RNA, Ribosomal, 16S/genetics
10.
Inflamm Bowel Dis ; 27(8): 1302-1315, 2021 07 27.
Article in English | MEDLINE | ID: mdl-33501933

ABSTRACT

BACKGROUND: A dysfunctional gut epithelial barrier allows the augmented permeation of endotoxins, luminal antigens, and bacteria into the bloodstream, causing disease. The maintenance of gut epithelial barrier integrity may be regulated by multiple factors. Herein we analyze the role of leucine-rich repeat-containing protein 19 (LRRC19) in regulating the permeability of the gut epithelial barrier. METHODS: We utilized Lrrc19 knockout (KO) mice and clinical samples through transmission electron, intestinal permeability assay, Western blot, and immunofluorescence staining to characterize the role of LRRC19 in the permeability of the gut epithelial barrier. RESULTS: We found that LRRC19, which is expressed in gut epithelial cells, impairs gut barrier function. Transmission electron micrographs revealed a tighter junction and narrower gaps in the colon epithelium cells in LRRC19 KO mice. There were lower levels of serum lipopolysaccharide and 4 kDa-fluorescein isothiocyanate-dextran after gavage in LRRC19 KO mice than in wild-type mice. We found that LRRC19 could reduce the expression of zonula occludens (ZO)-1, ZO-3, and occludin in the colonic epithelial cells. The decreased expression of ZO-1, ZO-3, and occludin was dependent on degrading protein kinase C (PKC) ζ and PKCι/λ through K48 ubiquitination by LRRC19. The expression of LRRC19 was also negatively correlated with ZO-1, ZO-3, occludin, PKCζ, and PKCι/λ in human colorectal cancers. CONCLUSIONS: The protein LRRC19 can promote the permeability of the gut epithelial barrier through degrading PKC ζ and PKCι/λ to reduce the expression of ZO-1, ZO-3, and occludin.


Subject(s)
Intestinal Mucosa/metabolism , Occludin/metabolism , Protein Kinase C , Receptors, Cell Surface/metabolism , Tight Junctions , Zonula Occludens-1 Protein/metabolism , Animals , Mice , Mice, Knockout , Permeability , Protein Kinase C/metabolism , Zonula Occludens Proteins
11.
Int Immunopharmacol ; 89(Pt A): 107030, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33045573

ABSTRACT

Macrophages, which can secret various inflammation mediators, have an essential role in tumor growth and metastasis. However, the mechanism(s) to regulate the production of inflammation mediator is not completely clear. Here we found that TRIM 59 could inhibit the production of NO and the expression of inducible nitric oxide synthase (iNOS), cytochrome c oxidase subunit2 (COX2) and TNFα. TRIM59 mediated suppression on nitric oxide (NO) production is through inhibiting the activation of JAK2-STAT1 signal pathway. In response to LPS, TRIM59 in macrophages was translocated from cytoplasm to nucleus and directly bound with STAT1. During this process, TRIM59 could recruit much more PIAS1 to bind with STAT1 to suppress the activation of STAT1. Finally, TRIM59 modified macrophages could promote tumor growth. Thus, TRIM59 mediated suppression on NO production by promoting the binding of PIAS1 and STAT1 in macrophages may regulate tumor growth.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Nitric Oxide/metabolism , Protein Inhibitors of Activated STAT/metabolism , STAT1 Transcription Factor/metabolism , Tripartite Motif Proteins/metabolism , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Down-Regulation , Intracellular Signaling Peptides and Proteins/genetics , Janus Kinase 2/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/transplantation , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Protein Binding , Protein Inhibitors of Activated STAT/genetics , RAW 264.7 Cells , Tripartite Motif Proteins/genetics , Tumor Burden , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/transplantation
12.
Cell Death Differ ; 26(7): 1181-1194, 2019 07.
Article in English | MEDLINE | ID: mdl-30464224

ABSTRACT

Pre-mRNA splicing is a fundamental process that plays a considerable role in generating protein diversity. Pre-mRNA splicing is also the key to the pathology of numerous diseases, especially cancers. In this review, we discuss how aberrant splicing isoforms precisely regulate three basic functional aspects in cancer: proliferation, metastasis and apoptosis. Importantly, clinical function of aberrant splicing isoforms is also discussed, in particular concerning drug resistance and radiosensitivity. Furthermore, this review discusses emerging strategies how to modulate pathologic aberrant splicing isoforms, which are attractive, novel therapeutic agents in cancer. Last we outline current and future directions of isoforms diagnostic methodologies reported so far in cancer. Thus, it is highlighting significance of aberrant splicing isoforms as markers for cancer and as targets for cancer therapy.


Subject(s)
Neoplasms/genetics , RNA Precursors/genetics , RNA Splicing/genetics , RNA, Messenger , Animals , Humans , Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Artif Cells Nanomed Biotechnol ; 47(1): 1273-1280, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30963795

ABSTRACT

Pladienolide B is a potent cancer cell growth inhibitor that targets the SF3b1 subunit of the spliceosome. There is considerable interest in the compound as a tool to study SF3b1 function in cancer. However, so far little information is available on the molecular mechanism of SF3b1 eliciting apoptosis in cancer cells. Here, we investigated the molecular mechanism of SF3b1 eliciting apoptosis in human cervical carcinoma cells. We demonstrated that inhibition of SF3b1 by pladienolide B inhibited proliferation of HeLa cells at low nanomolar concentrations in a dose- and time-dependent manner. It also induced G2/M phase arrest and significant rise of apoptotic cells. Moreover, it is indicated that inhibition of SF3b1 by pladienolide B induced Tap73/ΔNp73 expression and consequently down-regulated Bax/Bcl-2 ratio, cytochrome c release and caspase-3 expression. Thus, our results showed that SF3b1 plays a pivotal role in cycle arrest, apoptosis induction, and p73 splicing in human cervical carcinoma cells, suggesting that SF3b1 could be used as a potential candidate for cervical cancer therapy.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Epoxy Compounds/pharmacology , Macrolides/pharmacology , Phosphoproteins/antagonists & inhibitors , RNA Splicing Factors/antagonists & inhibitors , RNA Splicing/drug effects , Tumor Protein p73/genetics , Uterine Cervical Neoplasms/pathology , Cell Survival/drug effects , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Mitochondria/drug effects , Mitochondria/pathology
14.
Syst Biol Reprod Med ; 65(1): 61-70, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30452285

ABSTRACT

Male infertility affects approximately 7% of the male population. In about 40% of affected patients, the etiology remains unknown. Here, we report the cases of two infertile brothers who have a uniquely prevalent sperm phenotype with completely amorphous sperm heads. To investigate the mechanisms of familial teratozoospermia with amorphous sperm heads, chromatin condensation was assessed by aniline blue staining, western blot, sperm chromatin structure assay and atomic force microscopy in both the two brothers and 40 control fertile donors. Our results showed an abnormal condensation of chromatin with amorphous headed sperm. We suggest that abnormal chromatin condensation which was induced by disturbances in the process of histone-protamine replacement may be a possible cause of familial teratozoospermia with amorphous head, and the elasticity of sperm nuclei could be a new index to assess sperm quality. Additionally, for the first time, the current study provided a new biomechanics strategy for evaluating pathological sperm contributes to our understanding of teratozoospermia.Abbreviations: SCSA: sperm chromatin structure assay; AFM: atomic force microscopy; ICSI: intracytoplasmic sperm injection; HDS: high DNA stainability; DFI: DNA fragmentation index; PBS: phosphate-buffered saline; DTT: dithiothreitol; FITC: fluorescein isothiocyanate; DAPI: 4',6-diamidino-2-pheneylindole; SSC: standard saline citrate.


Subject(s)
Sperm Head/pathology , Teratozoospermia/pathology , Adult , DNA Packaging , Humans , Male , Young Adult
15.
Artif Cells Nanomed Biotechnol ; 46(sup3): S564-S570, 2018.
Article in English | MEDLINE | ID: mdl-30431368

ABSTRACT

Exosomes are nanoscale vesicles shed from all cell types and play a major role in communication and transportation of materials between cells due to their ability to transfer proteins and nucleic acids from one cell to another. Analogous in size and function to synthetic nanoparticles, exosomes offer many advantages, rendering them the most promising candidates for targeted drug or gene delivery vehicles. Exosomes can also induce chemoresistance or radioresistance of tumor cells. Studies about the related mechanisms help overcome cancer therapy resistance to some extent. In this review, we focus on the application of exosomes as nanocarriers and the current status of the application of exosomes to cancer therapy.


Subject(s)
Drug Carriers/therapeutic use , Exosomes , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Humans , Neoplasms/pathology
16.
Mutat Res ; 793-794: 41-50, 2016.
Article in English | MEDLINE | ID: mdl-27837686

ABSTRACT

The inhibitory effects of carbon monoxide (CO), generated by Ru(CO)3Cl-glycinate [CO-releasing molecule (CORM-3)], on developmental toxicity in zebrafish embryos induced by ionizing radiation with different linear energy transfer (LET) were studied. Zebrafish embryos at 5h post-fertilization were irradiated with X-ray (low-LET) and carbon-ion (high-LET) with or without pretreatment of CORM-3 1h before irradiation. CORM-3 pre-treatment showed a significant inhibitory effect on X-ray irradiation-induced developmental toxicity, but had little effect on carbon-ion irradiation-induced developmental toxicity. X-ray irradiation-induced significant increase in ROS levels and cell apoptosis could be modified by CORM-3 pretreatment. However, embryos exposed to carbon-ion irradiation showed significantly increase of cell apoptosis without obvious ROS generation, which could not be attenuated by CORM-3 pretreatment. CORM-3 could inhibit apoptosis induced by ionizing radiation with low-LET as an effective ROS scavenger. The expression of pro-apoptotic genes increased significantly after X-ray irradiation, but increased expression was reduced markedly when CORM-3 was applied before irradiation. Moreover, the protein levels of P53 and γ-H2AX increased markedly after X-ray irradiation, which could be modified by the presence of CORM-3. The protective effect of CORM-3 on X-ray irradiation occurred mainly by suppressing ROS generation and DNA damage, and thus inhibiting the activation of P53 and the mitochondrial apoptotic pathway, leading to the attenuation of cell apoptosis and consequently alleviating X-ray irradiation-induced developmental toxicity at lethal and sub-lethal levels.


Subject(s)
Carbon/adverse effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/radiation effects , Heavy Ions/adverse effects , Organometallic Compounds/pharmacology , X-Rays/adverse effects , Animals , DNA Damage/drug effects , DNA Damage/radiation effects , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/radiation effects , Radiation, Ionizing , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL