Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cancer Sci ; 114(4): 1229-1239, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36601880

ABSTRACT

Immune checkpoint inhibitors (ICIs) have become important treatment strategies, yet responses vary among patients and predictive biomarkers are urgently needed. Mutations in KMT2C and KMT2D lead to increased levels of genomic instability. Therefore, we aimed to examine whether KMT2C/D mutations might be a predictor of immunotherapeutic efficacy. Here, we investigated the associations of KMT2C/D loss-of-function (LOF) variants with tumor mutation burden (TMB), MSI-H, PD-L1 expression, the levels of tumor-infiltrating leukocytes (TILs), and clinical response to ICIs. It was found that KMT2C/D LOF variants were associated with higher TMB. Compared with the non-LOF group, the proportion of patients with MSI-H tumors was larger in the LOF group. PD-L1 expression was higher in the LOF group only for colorectal cancer in both the Chinese and The Cancer Genome Atlas cohorts. Importantly, KMT2C/D LOF variants were associated with decreased regulatory T cells and increased levels of CD8+ T cells, activated NK cells, M1 macrophages, and M2 macrophages in colorectal cancer. However, there was no significant association between KMT2C/D LOF and TILs levels in other cancer types. Consistently, the results showed that KMT2C/D LOF variants were associated with prolonged overall survival only in colorectal cancer (p = 0.0485). We also presented that patients with KMT2C/D LOF mutations exhibited a better clinical response to anti-PD-1 therapy in a Chinese colorectal cancer cohort (p = 0.002). Taken together, these results suggested that KMT2C/D LOF variants could be a useful predictor for ICIs efficacy in colorectal cancer. In addition, the predictive value of KMT2C/D LOF variants was consistent with their association with TILs levels.


Subject(s)
B7-H1 Antigen , Colorectal Neoplasms , Humans , B7-H1 Antigen/genetics , Immune Checkpoint Inhibitors/therapeutic use , CD8-Positive T-Lymphocytes , Mutation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Biomarkers, Tumor/genetics , Microsatellite Instability
2.
Transl Lung Cancer Res ; 10(11): 4235-4249, 2021 Nov.
Article in English | MEDLINE | ID: mdl-35004253

ABSTRACT

BACKGROUND: This study aimed to verify the feasibility of human epidermal growth factor receptor-2 (HER2) amplification detection by digital polymerase chain reaction (dPCR) in non-small cell lung cancer (NSCLC) patients and explore whether HER2 amplification could be detected in circulating tumor DNA (ctDNA) by dPCR. METHODS: A total of 112 fresh biopsy tissues and 88 blood samples from NSCLC patients were collected. The serum ctDNA was obtained from blood samples. The copy number of the HER2 gene was evaluated by dPCR and next-generation sequencing (NGS). The sensitivity/specificity and survival analysis were performed by the receiver operating characteristic (ROC) curve. The survival analysis was performed by Kaplan-Meier (KM) curve and univariate Cox regression analysis was also conducted. RESULTS: ROC analysis showed a good prediction result for HER2 amplification in blood samples by dPCR. The survival analysis showed that the median overall survival (OS) in the HER2 negative group detected by blood dPCR was significantly different from the positive group. The results of multivariate Cox regression were the same as those of survival analysis. CONCLUSIONS: Blood dPCR might be a potential method to detect HER2 amplification in NSCLC. Amplification of the HER2 gene detected by dPCR was correlated with OS in NSCLC.

3.
J Clin Pathol ; 74(5): 314-320, 2021 May.
Article in English | MEDLINE | ID: mdl-32817175

ABSTRACT

BACKGROUND: The absence of high-quality next-generation sequencing (NGS) reference material (RM) has impeded the clinical use of liquid biopsies with plasma cell-free DNA (cfDNA) in China. OBJECTIVE: This study aimed to develop a national RM panel for external quality assessment and performance evaluation during kit registration of non-small-cell lung cancer (NSCLC)-related Kirsten rat sarcoma viral oncogene (KRAS)/neuroblastoma ras oncogene (NRAS)/epidermal growth factor receptor (EGFR)/B-type Raf kinase (BRAF)/mesenchymal-epithelial transition factor (MET) genetic assays using plasma circulating tumor DNA (ctDNA). METHODS: Mutation cell lines detected by NGS and validated by Sanger sequencing were selected to establish the RM. Cell line genomic DNA was sheared and used to spike basal plasma cfDNA at 10% concentration. Then, the calibration accuracy was determined by four sequencing platforms. Average values were adopted and diluted to 0.1%, 0.3%, 1% and 3% concentrations with basal plasma as the RM panel. Then, five manufacturers were invited to evaluate the performance of the RM panel. RESULTS: 20 cell lines with 23 clinically important mutations were selected, including six mutations in KRAS, two mutations in NRAS, three in BRAF, four in phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), six in EGFR, one EGFR Gain (4-5 copy) and one MET Gain (2-5 copy). The RM panel consisted of 87 samples, including these 21 mutations at four concentrations (0.1%, 0.3%, 1% and 3%), one MET gain, one EGFR gain and one wild type. The detection rate was 100% for the 3%, 1% and 0.3% samples at all five companies. For the 0.1% concentration, 15 samples had inconsistent results, but at least three companies had correct results for each mutation. CONCLUSION: RM for a KRAS/NRAS/EGFR/BRAF/MET mutation panel for plasma ctDNA was developed, which will be essential for quality control of the performance of independent laboratories.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Circulating Tumor DNA/genetics , DNA Mutational Analysis/standards , GTP Phosphohydrolases/genetics , High-Throughput Nucleotide Sequencing/standards , Lung Neoplasms/genetics , Membrane Proteins/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Adult , Beijing , Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/diagnosis , Cell Line, Tumor , Circulating Tumor DNA/blood , ErbB Receptors/blood , ErbB Receptors/genetics , Female , GTP Phosphohydrolases/blood , Humans , Liquid Biopsy/standards , Lung Neoplasms/blood , Lung Neoplasms/diagnosis , Male , Membrane Proteins/blood , Middle Aged , Predictive Value of Tests , Proto-Oncogene Proteins B-raf/blood , Proto-Oncogene Proteins c-met/blood , Proto-Oncogene Proteins p21(ras)/blood , Reference Standards , Young Adult
4.
Front Oncol ; 10: 597486, 2020.
Article in English | MEDLINE | ID: mdl-33634022

ABSTRACT

BACKGROUND: Conventional clinical detection methods such as CT, urine cytology, and ureteroscopy display low sensitivity and/or are invasive in the diagnosis of upper tract urinary carcinoma (UTUC), a factor precluding their use. Previous studies on urine biopsy have not shown satisfactory sensitivity and specificity in the application of both gene mutation or gene methylation panels. Therefore, these unfavorable factors call for an urgent need for a sensitive and non-invasive method for the diagnosis of UTUC. METHODS: In this study, a total of 161 hematuria patients were enrolled with (n = 69) or without (n = 92) UTUC. High-throughput sequencing of 17 genes and methylation analysis for ONECUT2 CpG sites were combined as a liquid biopsy test panel. Further, a logistic regression prediction model that contained several significant features was used to evaluate the risk of UTUC in these patients. RESULTS: In total, 86 UTUC- and 64 UTUC+ case samples were enrolled for the analysis. A logistic regression analysis of significant features including age, the mutation status of TERT promoter, and ONECUT2 methylation level resulted in an optimal model with a sensitivity of 94.0%, a specificity of 93.1%, the positive predictive value of 92.2% and a negative predictive value of 94.7%. Notably, the area under the curve (AUC) was 0.957 in the training dataset while internal validation produced an AUC of 0.962. It is worth noting that during follow-up, a patient diagnosed with ureteral inflammation at the time of diagnosis exhibiting both positive mutation and methylation test results was diagnosed with ureteral carcinoma 17 months after his enrollment. CONCLUSION: This work utilized the epigenetic biomarker ONECUT2 for the first time in the detection of UTUC and discovered its superior performance. To improve its sensitivity, we combined the biomarker with high-throughput sequencing of 17 genes test. It was found that the selected logistic regression model diagnosed with ureteral cancer can evaluate upper tract urinary carcinoma risk of patients with hematuria and outperform other existing panels in providing clinical recommendations for the diagnosis of UTUC. Moreover, its high negative predictive value is conducive to rule to exclude patients without UTUC.

5.
Gene ; 704: 103-112, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-30974197

ABSTRACT

Lynch syndrome (LS) is a common cancer syndrome that is inherited in an autosomal dominant manner. Its pathogenesis is thought to be closely related to germline mutations of mismatch repair (MMR) genes such as the MLH1, MSH2, PMS2 and MSH6 genes. This study identifies a Chinese family with LS clinically diagnosed according to the Amsterdam II criteria. In these patients, immuno-histochemical staining showed negative MSH6 expressions but positive MLH1, MSH2, and PMS2 expressions. In order to further explore the molecular biology of this LS family, we used targeted next-generation sequencing (NGS) and Multiplex ligation dependent probe amplification (MLPA) to identify the mutation and verify the authenticity of the mutation in 15 family members. For NGS, two panels have been used, one is of MLH1, MSH2, PMS2 and MSH6 genes, the other one is of 139 cancer genetic susceptibility genes. And for the large deletions/duplications can also be identified by NGS panel, an adjusted data analysis strategy of NGS has been used. As a result, we identified a novel heterozygous large deletion in MSH6 gene that was found to be co-segregated among affected family members. This deletion results in the loss of a 3246 bp-sized fragment in MSH6 gene exons 5-9 which represents the coding regions of the MSH6 ATPase domain. This novel mutation has yet to be documented in the International Society for Gastrointestinal Hereditary Tumours (InSiGHT) database. This mutation resulted in MSH6 protein losing gene mismatch repair function, and further caused the microsatellite instable. We speculate that this mutation may significantly impact MMR function through impaired ATP domain function. Theoretically, this proband would likely benefit from PD-1 immune check-point blockade therapy, but conversely, we observed that tumors appeared to rapidly progress after 4 sessions of anti-PD-1 treatment. Further studies to validate the effectiveness of anti-PD-1 treatments in carriers of this mutation are necessary.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA-Binding Proteins/genetics , Germ-Line Mutation , Sequence Deletion , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Asian People/genetics , China , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Mismatch Repair/genetics , Family , Female , Genetic Testing , Heterozygote , Humans , Male , Microsatellite Instability , Pedigree
6.
Am J Physiol Endocrinol Metab ; 307(12): E1131-43, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25336526

ABSTRACT

Wilms' tumor 1 (Wt1) is a tumor suppressor gene encoding ∼24 zinc finger transcription factors. In the mammalian testis, Wt1 is expressed mostly by Sertoli cells (SCs) involved in testis development, spermatogenesis, and adult Leydig cell (ALC) steroidogenesis. Global knockout (KO) of Wt1 is lethal in mice due to defects in embryogenesis. Herein, we showed that Wt1 is involved in regulating fetal Leydig cell (FLC) degeneration and ALC differentiation during testicular development. Using Wt1(-/flox);Amh-Cre mice that specifically deleted Wt1 in the SC vs. age-matched wild-type (WT) controls, FLC-like-clusters were found in Wt1-deficient testes that remained mitotically active from postnatal day 1 (P1) to P56, and no ALC was detected at these ages. Leydig cells in mutant adult testes displayed morphological features of FLC. Also, FLC-like cells in adult mutant testes had reduced expression in ALC-associated genes Ptgds, Sult1e1, Vcam1, Hsd11b1, Hsd3b6, and Hsd17b3 but high expression of FLC-associated genes Thbs2 and Hsd3b1. Whereas serum LH and testosterone level in mutant mice were not different from controls, intratesticular testosterone level was significantly reduced. Deletion of Wt1 gene also perturbed the expression of steroidogenic enzymes Star, P450c17, Hsd3b6, Hsd3b1, Hsd17b1, and Hsd17b3. FLCs in adult mutant testes failed to convert androstenedione to testosterone due to a lack of Hsd17b3, and this defect was rescued by coculturing with fetal SCs. In summary, FLC-like cells in mutant testes are putative FLCs that remain mitotically active in adult mice, illustrating that Wt1 dictates the fate of FLC and ALC during postnatal testis development.


Subject(s)
Cell Differentiation/genetics , Leydig Cells/physiology , Testis/embryology , Testis/growth & development , WT1 Proteins/physiology , Animals , Embryo, Mammalian , Fetus/embryology , Fetus/physiology , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sertoli Cells/physiology , Testis/cytology
7.
Reproduction ; 147(1): 45-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24129152

ABSTRACT

Spermatogenesis is a complex process involving the regulation of multiple cell types. As the only somatic cell type in the seminiferous tubules, Sertoli cells are essential for spermatogenesis throughout the spermatogenic cycle. The Wilms tumor gene, Wt1, is specifically expressed in the Sertoli cells of the mouse testes. In this study, we demonstrated that Wt1 is required for germ cell differentiation in the developing mouse testes. At 10 days post partum, Wt1-deficient testes exhibited clear meiotic arrest and undifferentiated spermatogonia accumulation in the seminiferous tubules. In addition, the expression of claudin11, a marker and indispensable component of Sertoli cell integrity, was impaired in Wt1(-/flox); Cre-ER(TM) testes. This observation was confirmed in in vitro testis cultures. However, the basal membrane of the seminiferous tubules in Wt1-deficient testes was not affected. Based on these findings, we propose that Sertoli cells' status is affected in Wt1-deficient mice, resulting in spermatogenesis failure.


Subject(s)
Meiosis/physiology , Sertoli Cells/metabolism , Spermatogenesis/physiology , Spermatogonia/metabolism , WT1 Proteins/metabolism , Animals , Claudins/genetics , Claudins/metabolism , Male , Mice , Mice, Knockout , WT1 Proteins/genetics
8.
Biol Reprod ; 89(1): 12, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23759306

ABSTRACT

Scrotal hypothermia is essential for normal spermatogenesis, and temporal heat stress causes a reversible disruption of the blood-testis barrier (BTB). Previous studies have shown that AR expression in primary monkey Sertoli cells (SCs) was dramatically reduced after temporary heat treatment. However, the mechanisms underlying the heat-induced reversible disruption of the BTB, including whether it is directly regulated by the AR, remain largely unknown. In this study, we demonstrated that the AR acts upstream to regulate the heat-induced reversible change in the BTB in mice. When the AR was overexpressed in SCs using an adenovirus, the heat stress-induced down-regulation of BTB-associated proteins (Zonula occludens-1 (ZO-1), N-Cadherin, E-Cadherin, α-Catenin, and ß-Catenin) was partially rescued. AR knockdown by RNAi or treatment with flutamide (an AR antagonist) in SCs inhibited the recovery of BTB-associated protein expression after 43°C heat treatment for 30 min. The results of an in vivo AR antagonist injection experiment further showed that the recovery of BTB permeability induced by temporal heat stress was regulated by the AR. Furthermore, we observed that the co-localization and interactions of partitioning-defective protein (Par) 6-Par3-aPKC-Cdc42 polarity complex components were disrupted in both AR-knockdown and heat-induced SCs. AR overexpression in SCs prevented the disruption of these protein-protein interactions after heat treatment. AR knockdown or treatment with flutamide in SCs inhibited the restoration of these protein-protein interactions after heat treatment compared with heat treatment alone. Together, these results demonstrate that the AR plays a crucial role in the heat-induced reversible change in BTB via the Par polarity complex.

9.
BMC Biol ; 11: 22, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23497137

ABSTRACT

BACKGROUND: The directional migration and the following development of primordial germ cells (PGCs) during gonad formation are key steps for germline development. It has been proposed that the interaction between germ cells and genital ridge (GR) somatic cells plays essential roles in this process. However, the in vivo functional requirements of GR somatic cells in germ cell development are largely unknown. RESULTS: Wt1 mutation (Wt1(R394W/R394W)) results in GR agenesis through mitotic arrest of coelomic epitheliums. In this study, we employed the GR-deficient mouse model, Wt1(R394W/R394W), to investigate the roles of GR somatic cells in PGC migration and proliferation. We found that the number of PGCs was dramatically reduced in GR-deficient embryos at embryonic day (E) 11.5 and E12.5 due to decreased proliferation of PGCs, involving low levels of BMP signaling. In contrast, the germ cells in Wt1(R394W/R394W) embryos were still mitotically active at E13.5, while all the germ cells in control embryos underwent mitotic arrest at this stage. Strikingly, the directional migration of PGCs was not affected by the absence of GR somatic cells. Most of the PGCs reached the mesenchyme under the coelomic epithelium at E10.5 and no ectopic PGCs were noted in GR-deficient embryos. However, the precise positioning of PGCs was disrupted. CONCLUSIONS: Our work provides in vivo evidence that the proliferation of germ cells is precisely regulated by GR somatic cells during different stages of gonad development. GR somatic cells are probably dispensable for the directional migration of PGCs, but they are required for precise positioning of PGCs at the final step of migration.


Subject(s)
Cell Movement , Germ Cells/cytology , Gonads/cytology , Gonads/embryology , Animals , Cell Communication , Cell Count , Cell Proliferation , Chemokine CXCL12/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Epithelium/embryology , Epithelium/metabolism , Female , Integrin beta1/metabolism , Male , Mice , Mitosis , Mutation/genetics , Sex Determination Processes , Stem Cell Factor/metabolism , WT1 Proteins/genetics
10.
Biol Reprod ; 88(3): 56, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23325811

ABSTRACT

Wt1 is specifically expressed in Sertoli cells in the developing testis. A previous study has demonstrated that Wt1 plays a critical role in maintaining the integrity of testicular cords. However, the underlying mechanism is unclear. In this study, we found that the laminin-positive basal lamina lining the testicular cords was fragmented and completely absent in some areas of Wt1(-/flox); Amh-Cre testes, indicating that the testicular cord disruption can be attributed to the breakdown of the basement membrane. To explore the molecular mechanism underlying this effect, we examined the expression of cell adhesion molecules (CAMs) and testicular cord basal lamina components by real-time RT-PCR, Western blotting, and immunostaining. Compared with control testes, the expression of CAMs (such as E-cadherin, N-cadherin, claudin11, occludin, beta-catenin, and ZO-1) was not obviously altered in Wt1(-/flox); Amh-Cre testes. However, the mRNA level of Col4a1 and Col4a2 was significantly decreased in Wt1-deficient testes. Immunostaining assays further confirmed that the collagen IV protein levels were dramatically reduced in Wt1(-/flox); Amh-Cre testes. Moreover, luciferase and point mutation analyses revealed that the Col4a1 and Col4a2 promoters were additively transactivated by WT1 and SOX9. Given this finding and previous results showing that SOX9 expression declines rapidly after Wt1 deletion, we conclude that the loss of Wt1 in Sertoli cells results in the downregulation of the important basal lamina component, which in turn causes the breakdown of the basal lamina and subsequent testicular cord disruption.


Subject(s)
Collagen Type IV/metabolism , Genes, Wilms Tumor , Spermatic Cord/embryology , Testis/metabolism , Animals , Basement Membrane/physiology , Cell Adhesion Molecules/metabolism , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , SOX9 Transcription Factor/metabolism , Testis/embryology , Transcriptional Activation
11.
PLoS One ; 8(1): e53140, 2013.
Article in English | MEDLINE | ID: mdl-23326390

ABSTRACT

Wt1 encodes a zinc finger nuclear transcriptional factor, which is specifically expressed in testicular Sertoli cells and knockdown of Wt1 in Sertoli cells causes male mice subfertility. However, the underlying mechanism is still unclear. In this study, we found that expression of inhibin-α is significantly reduced in Wt1-deficient Sertoli cells. Luciferase assays using the inhibin-α promoter indicated that the inhibin-α promoter is transactivated by the Wt1 A, and B isoforms (-KTS), but not the C, and D isoforms (+KTS). Analysis of the Wt1 responsive element of the inhibin-α promoter region using site-directed mutagenesis showed that the nucleotides between -58 and -49 are essential for Wt1-dependent transactivation of the inhibin-α promoter. ChIP assays indicated that Wt1 directly interacts with the inhibin-α promoter. In addition, the inhibin-α promoter is activated synergistically by Wt1 and Sf1. Mutation of the ligand binding domain (LBD) of Sf1 (residues 235-238) completely abolished the synergistic action between Wt1 and Sf1, but did not affect the physical interaction between these two proteins, suggesting that other factor(s) may also be involved in the regulation of inhibin-α in Sertoli cells. Further studies demonstrated that ß-catenin enhances the synergistic activation of Wt1 and Sf1 on the inhibin-α promoter. Given the fact that inhibin-α, a subunit of inhibin, is known to be involved in the regulation of spermatogenesis and testicular steroidogenesis, this study reveals a new regulatory mechanism of inhibin-α in Sertoli cells and also sheds light on the physiological functions of Wt1 in gonad development and spermatogenesis.


Subject(s)
Gene Expression Regulation , Inhibins/genetics , Sertoli Cells/metabolism , Steroidogenic Factor 1/genetics , WT1 Proteins/genetics , Animals , Base Sequence , Binding Sites/genetics , Blotting, Western , Cell Line , Cells, Cultured , Female , Inhibins/metabolism , Male , Mice , Mice, Knockout , Mutation , Promoter Regions, Genetic/genetics , Protein Isoforms/genetics , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroidogenic Factor 1/metabolism , Transcriptional Activation , WT1 Proteins/metabolism , Wnt Signaling Pathway/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...