GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (11)
  • Yang, Fan  (11)
Material
Publisher
  • American Association for Cancer Research (AACR)  (11)
Language
Years
Subjects(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 618-618
    Abstract: A portion of stage I non-small cell lung cancer (NSCLC) patients who have received R0 resection experience unexpected tumor recurrence during follow up. Some of them even recurred within 12 months after surgery, which is unable to predict by any clinical characteristics. Few study investigates the molecular features of such short-term recurrent tumors. Multiomics analysis were performed on 81 stage I NSCLC patients who had received R0 resection, based on deep whole-exome sequencing (average depth & gt;500x) on multi-region 239 tumor tissue samples and 81 matched-adjacent normal specimens, as well as RNA sequencing and plasma targeted circulating tumor DNA detection. Dynamic dimension comparison of genomic alteration features between tumor recurrence and non-recurrence patients was made according to 60-months median time of follow up. Published WES datasets of Stage I NSCLCs of 131 East Asian (EAS LUADs), 62 European (TRACERx) and 277 American patients (TCGA) were extracted as validation. 60.5% patients were non-smokers. Lower chromosomal instability was found in our cohort characterized by less trunk CNVs and lower CNV coverage on genome as compared to the European cohort. Age-related signature mutations significantly accumulated in recurrence-free tumors. Somatic mutations occurred in DNA damage repair pathways were more likely to cause tumor recurrence (p=0.04), particularly in homologous recombination (HRR) (p=0.017) and translesion synthesis (TLS) (p=0.009) pathways. Dynamic analysis discovered the essential genes in Fanconi anemia (FA), TLS and HRR pathways that coordinate together for DNA interstrand crosslink (ICL) recognition and faithful repair of DNA double strand breaks (DSBs) during DNA replication stress frequently mutated in early tumor recurrence patients (n=7/11, 63%). Twenty-two focal CNV-driver genes, including cell proliferation-related RECQL4 (8q24.3), PIK3CA (3q26.32), BCL9 (1q21.2), TBL1XR1 (3q26.32) frequently amplified in recurrent tumors, as well as BCL3 and CBLC (19q13.31-32) in the early recurrent tumors. RNA-seq data demonstrated cell cycle pathway significantly up-regulated in recurrent tumors. More TMB, CNV fraction and genome doubling events in FA-TLS-HRR alteration patients verified genomic instability caused by dysfunction of cell cycle control and ICL-DSB repair contributing to early tumor recurrence. Integrative model cross clinical and molecular features stratified stage I patient prognosis in both our cohort and European cohort, in which and high risk patients were more detected by circulating tumor DNA. This is the first study to dynamically investigate the genomic features specifically in stage I lung NSCLC according to the precise tumor recurrence time, which provides important clues to postoperative treatment strategy and drug target research. Citation Format: Kezhong Chen, Airong Yang, Shuangxiu Wu, Yuntao Nie, Haifeng Shen, Jian Bai, Lin Wu, Fan Yang, Jun Wang. Spatio-temporal multiomics analysis reveals distinct molecular features in recurrent stage I non-small cell lung cancers after R0 tumor resection [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 618.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5916-5916
    Abstract: Background: Patient-specific flexible gene panels designed based on whole-exome sequencing (WES) of resected tumor tissues is a promising strategy for ctDNA-based detection of molecular residual disease (MRD) in early-stage NSCLC. Flexible gene panels could potentially overcome the limitations of fixed panels by incorporating more unique genomic regions that might be absent in fixed panels; however, no study has reported a head-to-head comparison of these two approaches in postoperative disease monitoring. In this study, we investigated the clinical utility of a novel Patient-specific pROgnostic and Potential tHErapeutic marker Tracking (PROPHET) tumor-informed ctDNA assay. Using the same set of longitudinal blood samples, we further compared the performance of PROPHET assay with tumor-informed (TI) and tumor-naïve (TN) fixed panels for predicting MRD and prognosis in surgical NSCLC patients. Methods: Fifty-three patients with stage I-III resected NSCLC from the MEDAL study (NCT03634826) with adequate samples and median follow-up of 647 days were analyzed. Matched surgical tumor tissue and blood samples collected at various time points, including before surgery (baseline), 3-days (B) and 1-month (C) postoperative time points before any adjuvant therapy and subsequent follow-up time points (F) were analyzed. PROPHET assay involved four major steps: identify somatic mutations using WES, customized design of a patient-specific panel consisting of 50 single nucleotide variants, ultra-deep unique molecular-identifier-based next-generation sequencing (UMI-NGS) of serial blood samples using the patient-specific panel, and MRD risk prediction. Fixed panel assay of serial blood samples was performed using UMI-NGS with 168 gene panel spanning 273 kb of human genome. Results: At 1-month post-surgery, PROPHET assay accurately predicted MRD-positive cases among relapsed patients (50%, 13/26), whereas all disease-free patients were MRD-negative (100%, 21/21). Three-year prognostication with PROPHET assay at B+C yielded higher sensitivity (59% vs 26% vs 22%), negative predictive value (66% vs 51% vs 50%), and hazard ratio (7.15, 95%CI [3.2-15.9] vs 4.48 [1.9-10.9] vs 5.58 [2.1-14.7]) as compared with TI and TN fixed panel assays. Disease monitoring using PROPHET assay at B/C/F accurately predicted the MRD risk in 70% (21/30) of relapsed patients at a median lead time of 318 days (range: 20-751), whereas TI assay predicted 43% (13/30) at 282 days (range 20-716) and TN assay predicted 37% (11/30) at 282 days (range: 20-634). Conclusion: Patient-specific tumor-informed ctDNA-based postoperative monitoring enables risk stratification at early postoperative settings better than fixed panel, which paves an alternative strategy in the individualized management of surgical NSCLC patients. Citation Format: Kezhong Chen, Haifeng Shen, Shuailai Wu, Pengfei Zhu, Chenyang Wang, Analyn Lizaso, Guannan Kang, Yang Wang, Juan Lv, Shuai Fang, Wenjun Wu, Fujun Qiu, Yuan Sun, Qiang Lu, Heng Zhao, Shannon Chuai, Fan Yang, Zhihong Zhang. Tumor-informed patient-specific panel outperforms tumor-naïve and tumor-informed fixed panel for circulating tumor DNA (ctDNA)-based postoperative monitoring of non-small cell lung cancer (NSCLC) [abstract]. In: Proceedings of the Amer ican Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5916.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 864-864
    Abstract: Persistent Jak/Stat3 signal transduction plays a crucial role in tumorigenesis and immune development. Activated Jak/Stat3 signaling has been validated as a promising molecular target for cancer therapeutics discovery and development. Berbamine (BBM), a natural bis-benzylisoquinoline alkaloid, was identified from the traditional Chinese herbal medicine Berberis amurensis used for treatment of cancer patients. While BBM has been shown to have potent antitumor activities with low toxicity in various cancer types, the molecular mechanism of action of BBM remains largely unknown. Here, we determine the antitumor activities of thirteen synthetic berbamine derivatives (BBMDs) against human solid tumor cells. BBMD3, which is the most potent in this series of novel BBMDs, exhibits over 6-fold increase in biological activity compared to natural BBM. Moreover, BBMD3, directly inhibits Jak2 autophosphorylation kinase activity in vitro with IC50 = 0.69 μM. Autophosphorylation of Jak2 kinase at Tyr1007/1008 sites also was strongly inhibited in the range of 1 μM to 5 μM of BBMD3 in human melanoma cells 4 h after treatment. Following inhibition of autophosphorylation of Jak2, BBMD3 blocked constitutive activation of downstream Stat3 signaling in melanoma cells. BBMD3 also down-regulated expression of Stat3 target proteins Mcl-1and Bcl-xL, associated with induction of apoptosis. In sum, our findings demonstrate that the novel berbamine derivative BBMD3 is an inhibitor of the Jak2/Stat3 signaling pathway, suggesting evidence for a molecular mechanism whereby BBMD3 exerts at least part of apoptosis of human melanoma cells. In addition, BBMD3 represents a promising lead compound for development of new therapeutics for cancer treatment. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 864. doi:1538-7445.AM2012-864
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 16, No. 9 ( 2017-09-01), p. 1806-1818
    Abstract: p53 deficiency, a frequent event in multiple kinds of malignancies, decreases the sensitivity of diverse targeted chemotherapeutics including the BCL-XL/BCL-2 inhibitor ABT-263. Loss of p53 function can activate mTOR complex 1 (mTORC1), which may make it a vulnerable target. Metformin has shown anti-neoplastic efficiency partially through suppressing mTORC1. However, it remains unknown whether mTORC1 activation confers ABT-263 resistance and whether metformin can overcome it in the p53-defective contexts. In this study, we for the first time demonstrated that metformin and ABT-263 synergistically elicited remarkable apoptosis through orchestrating the proapoptotic machineries in various p53-defective cancer cells. Mechanistic studies revealed that metformin sensitized ABT-263 via attenuating mTORC1-mediated cap-dependent translation of MCL-1 and survivin and weakening internal ribosome entry site (IRES)-dependent translation of XIAP. Meanwhile, ABT-263 sensitized metformin through disrupting the BCL-XL/BIM complex. However, metformin and ABT-263 had no synergistic killing effect in p53 wild-type (p53-WT) cancer cells because the cotreatment dramatically induced the senescence-associated secretory phenotype (SASP) in the presence of wild type p53, and SASP could aberrantly activate the AKT/ERK–mTORC1–4EBP1–MCL-1/survivin signaling axis. Blocking the axis using corresponding kinase inhibitors or neutralizing antibodies against different SASP components sensitized the cotreatment effect of metformin and ABT-263 in p53-WT cancer cells. The in vivo experiments showed that metformin and ABT-263 synergistically inhibited the growth of p53-defective (but not p53-WT) cancer cells in tumor xenograft nude mice. These results suggest that the combination of metformin and ABT-263 may be a novel targeted therapeutic strategy for p53-defective cancers. Mol Cancer Ther; 16(9); 1806–18. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 29, No. 8 ( 2023-04-14), p. 1484-1495
    Abstract: CD7 chimeric antigen receptor T (CAR-T) therapy has potent antitumor activity against relapsed/refractory (R/R) T-cell acute lymphoblastic leukemia/lymphoma (T-ALL/LBL), however, immune reconstitution after CAR-T remains largely unknown. Patients and Methods: An open-label phase I clinical trial (ChiCTR2200058969) was initiated to evaluate safety and efficacy of donor-derived CD7 CAR-T cells in 7 R/R T-ALL/LBL patients. CAR-T cells were detected by flow cytometry and PCR. Cytokine levels were quantified by cytometric bead arrays. Single-cell RNA sequencing (scRNA-seq) was adopted to profile immune reconstitution. Results: Optimal complete remission (CR) was 100% on day 28, and median followed-up time was 4 months. Leukopenia, thrombocytopenia, and neutropenia were observed in 6 patients, and infections occurred in 5 patients. Two patients died of serious infection and one died of a brain hemorrhage. CAR-T cells expanded efficiently in all patients. CD7+ T cells were eliminated in peripheral blood on day 11 after infusion, and CD7− T cells dramatically expanded in all patients. scRNA-seq suggested that immunologic activities of CD7− T cells were stronger than those of T cells before infusion due to higher expression levels of T-cell function-related pathways, and major characters of such CD7− T cells were activation of autoimmune-related pathways. Monocyte loss was found in 2 patients who died of serious infections, indicating the main cause of the infections after infusion. S100A8 and S100A9 were identified as potential relapse markers due to their notable upregulation in leukocyte lineage in relapsed patients versus non-relapse controls. Conclusions: Our data revealed cellular level dynamics of immune homeostasis of CD7 CAR-T therapy, which is valuable for optimizing the treatment of R/R T-ALL/LBL.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 696-696
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 696-696
    Abstract: Bone is one of the most common sites of cancer metastasis, affecting more than 70% of patients with advanced breast cancer and prostate cancer. Despite its prevalence, there are still no effective treatments for bone metastasis, leading to very poor survival rates and the mechanism by which certain cancer cells preferentially spread to the bones remains poorly understood. Thus, there is a critical need to develop in vitro experimental models that can mimic key aspects of bone metastasis development to elucidate driving mechanisms of this disease and expedite the discovery of novel drug candidates. To address this unmet need, here we report a spatially patterned, 3D in vitro model which mimics cancer metastasis to bone by incorporating interactions with multiple tissue types. The model is composed of an outer ring of tissue engineered bone derived from 3D osteogenic differentiation of human mesenchymal stem cells (hMSCs), and a center containing cancer cells. This interface between these two tissues mimics the way cancer cells invade into the bones through the bone marrow/bone interface and allows us to track the invasion of fluorescently labeled cancer cells into bone using confocal imaging. Cancer cells and hMSCs were also labeled with orthogonal luciferases (FireflyLuc and NanoLuc), allowing for simultaneous quantification of cancer cell proliferation and osteoblast survival using BLI. We tested the ability of this model to recapitulate known characteristics of in vivo bone metastases including tissue-specific invasion, cancer aggressiveness, cancer-induced bone resorption, and in vivo drug response. Using multiple established breast cancer (MDA-MB-231 and MCF-7) and prostate cancer (LNCaP and PC-3) cell lines, we demonstrate that such spatially patterned coculture models mimic the preferential invasion of cancer cells to bone, but not cartilage or muscle (negative controls) and the rate of invasion in this 3D model correlated with the level of cancer cell aggressiveness. To mimic the effect of cancer invasion on bone remodeling, we established a triculture model in which osteoclasts were seeded over the tissue engineered bone. Micro-computed tomography imaging validated that this model could recapitulate cancer-induced bone resorption, and the degree of bone resorption also correlated with cancer cell aggressiveness. Using parathyroid hormone as a model drug, we demonstrate this 3D model recapitulates drug responses consistent with what has been observed using in vivo mouse models. Such spatially patterned 3D models can provide a scalable platform for drug screening with substantially reduced time and cost compared to animal models. Furthermore, integrating these 3D cancer metastasis models with high-dimensional methods, such as single-cell RNAseq could expedite the discovery of novel druggable drivers of bone metastasis. Citation Format: Eva C. González Díaz, Michelle Tai, Callan E. Monette, Joy Wu, Fan Yang. Spatially patterned, 3D in vitro models of cancer metastasis to bone for elucidating key drivers of metastasis and drug discovery [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 696.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 6213-6213
    Abstract: B-cell acute lymphoblastic leukemia (B-ALL) is the most common pediatric malignancy. Although overall cure rate has exceeded 90%, the prognosis of high-risk B-ALL remains poor. Genome sequencing studies have revealed key genetic aberrations in B-ALL. But driver remains unclear in 8% of B-ALL for leukemogenesis, and 40% of high-risk B-ALL for drug resistance. Epigenetic mechanisms contributing to leukemia have recently been recognized, including histone modification and methylation. To date, a comprehensive landscape of chromatin accessibility in B-ALL is still lacking. Here, we performed Assays for Transposase Accessible Chromatin using sequencing (ATAC-seq) on a total of 61 high-risk B-ALLs treated at Shanghai Children’s Medical Center, including major B-ALL subtypes. We generated 144 high quality chromatin accessibility profiles from 79 tumors, including 18 diagnosis-relapse paired tumors as well as 11 diagnosis and 32 relapsed tumors. We observed significantly higher chromatin accessibility in B-ALL as compared to normal B-cells. Functional chromatin state annotation showed a median of 27.1% open chromatin regions (OCRs) in B-ALL were constituted from quiescent regions that were absent for known histone modifications, indicating unveiled role of quiescent regions in B-ALL. We further investigated the allelic imbalanced chromatin accessibility in 32 B-ALL with matched ATAC-seq and whole genome sequencing data. Unexpectedly, we found a median of 10.8% of OCRs showed imbalanced accessibility between two heterozygous alleles. Moreover, the adjacent imbalanced OCRs tend to be in the same topologically associating domain, suggesting the transcription regulation from chromatin accessibility was constrained within 3-D genome architecture. We next looked into the OCRs among B-ALL subtypes. Consistent with the regulatory role of chromatin accessibility on gene transcription, we found subtype specific pattern of OCRs in B-ALL, with open regions from enhancer and bivalent chromatin states showed more power in subtype discrimination. A total of 15516 out of 765788 (2.03%) peaks were identified as subtype specific OCRs. Motif analysis associated these regions to 212 transcription factors (TFs). Consistently, genes regulated by these TFs also exhibited subtype specific expression. Finally, we analyzed differences in OCRs between diagnosis (D) and relapse (R) in each B-ALL subtype. Only 2.44% D-R differential peaks were shared between any two subtypes, suggesting subtype specific role of chromatin accessibility in relapse. Potential target genes were further identified with quantitative trait loci analysis. With data from DepMap project, we verified that the expression of the target genes identified was associated with drug response. These data unveiled potential role of chromatin accessibility in high-risk B-ALL and the treatment response of this malignancy. Citation Format: Han Wang, Huiying Sun, Bilin Liang, Fang Zhang, Fan Yang, Bowen Cui, Lixia Ding, Ronghua Wang, Yanjing Tang, Jianan Rao, Wenting Hu, Shuang Zhao, Wenyan Wu, Benshang Li, Jingyan Tang, Shuhong Shen, Yu Liu. Chromatin accessibility landscape of pediatric high-risk B-cell acute lymphoblastic leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 6213.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 11 ( 2011-06-01), p. 3972-3979
    Abstract: STAT3 is persistently activated and contributes to malignant progression in various cancers. Janus activated kinases (JAK) phosphorylate STAT3 in response to stimulation by cytokines or growth factors. The STAT3 signaling pathway has been validated as a promising target for development of anticancer therapeutics. Small-molecule inhibitors of JAK/STAT3 signaling represent potential molecular-targeted cancer therapeutic agents. In this study, we investigated the role of JAK/STAT3 signaling in 6-bromoindirubin-3′-oxime (6BIO)-mediated growth inhibition of human melanoma cells and assessed 6BIO as a potential anticancer drug candidate. We found that 6BIO is a pan-JAK inhibitor that induces apoptosis of human melanoma cells. 6BIO directly inhibited JAK-family kinase activity, both in vitro and in cancer cells. Apoptosis of human melanoma cells induced by 6BIO was associated with reduced phosphorylation of JAKs and STAT3 in both dose- and time-dependent manners. Consistent with inhibition of STAT3 signaling, expression of the antiapoptotic protein Mcl-1 was downregulated. In contrast to the decreased levels of phosphorylation of JAKs and STAT3, phosphorylation levels of the Akt and mitogen-activated protein kinase (MAPK) signaling proteins were not inhibited in cells treated with 6BIO. Importantly, 6BIO suppressed tumor growth in vivo with low toxicity in a mouse xenograft model of melanoma. Taken together, these results show that 6BIO is a novel pan-JAK inhibitor that can selectively inhibit STAT3 signaling and induces tumor cell apoptosis. Our findings support further development of 6BIO as a potential anticancer therapeutic agent that targets JAK/STAT3 signaling in tumor cells. Cancer Res; 71(11); 3972–9. ©2011 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 14 ( 2022-07-18), p. 2576-2592
    Abstract: Recent work has made it clear that pericentriolar material (PCM), the matrix of proteins surrounding centrioles, contributes to most functions of centrosomes. Given the occurrence of centrosome amplification in most solid tumors and the unconventional survival of these tumor cells, it is tempting to hypothesize that gel-like mitotic PCM would cluster extra centrosomes to defend against mitotic errors and increase tumor cell survival. However, because PCM lacks an encompassing membrane, is highly dynamic, and is physically connected to centrioles, few methods can decode the components of this microscale matrix. In this study, we took advantage of differential labeling between two sets of APEX2-centrosome reactions to design a strategy for acquiring the PCM proteome in living undisturbed cells without synchronization treatment, which identified 392 PCM proteins. Localization of ubiquitination promotion proteins away from PCM was a predominant mechanism to maintain the large size of PCM for centrosome clustering during mitosis in cancer cells. Depletion of PCM gene kinesin family member 20A (KIF20A) caused centrosome clustering failure and apoptosis in cancer cells in vitro and in vivo. Thus, our study suggests a strategy for targeting a wide range of tumors exhibiting centrosome amplification and provides a proteomic resource for future mining of PCM proteins. Significance: This study identifies the proteome of pericentriolar material and reveals therapeutic vulnerabilities in tumors bearing centrosome amplification.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 18 ( 2019-09-15), p. 5525-5536
    Abstract: APOBEC3-UNG imbalance contributes to hepatitis B virus (HBV) inhibition and somatic mutations. We aimed to explore the associations between hepatocellular carcinoma (HCC) risk and genetic polymorphisms predisposing the imbalance. Experimental Design: Genetic polymorphisms at APOBEC3 promoter and UNG enhancer regions were genotyped in 5,621 participants using quantitative PCR. HBV mutations (nt.1600–nt.1945, nt.2848–nt.155) were determined by Sanger sequencing. Dual-luciferase reporter assay was applied to detect the transcriptional activity. Effects of APOBEC3B/UNG SNPs and expression levels on HCC prognosis were evaluated with a cohort of 400 patients with HCC and public databases, respectively. Results: APOBEC3B rs2267401-G allele and UNG rs3890995-C allele significantly increased HCC risk. rs2267401-G allele was significantly associated with the generation of APOBEC-signature HBV mutation whose frequency consecutively increased from asymptomatic HBV carriers to patients with HCC. Multiplicative interaction of rs2267401-G allele with rs3890995-C allele increased HCC risk, with an adjusted OR (95% confidence interval) of 1.90 (1.34–2.81). rs2267401 T-to-G and rs3890995 T-to-C conferred increased activities of APOBEC3B promoter and UNG enhancer, respectively. IL6 significantly increased APOBEC3B promoter activity and inhibited UNG enhancer activity, and these effects were more evident in those carrying rs2267401-G and rs3890995-C, respectively. APOBEC3B rs2267401-GG genotype, higher APOBEC3B expression, and higher APOBEC3B/UNG expression ratio in HCCs indicated poor prognosis. APOBEC-signature somatic mutation predicts poor prognosis in HBV-free HCCs rather than in HBV-positive ones. Conclusions: Polymorphic genotypes predisposing the APOBEC3B-UNG imbalance in IL6-presenting microenvironment promote HCC development, possibly via promoting the generation of high-risk HBV mutations. This can be transformed into specific prophylaxis of HBV-caused HCC.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...