1
|
Sun P, Zhao H, Cao L, Zhang T, Zhang H, Yang T, Zhao B, Jiang Y, Dong J, Chen T, Jiang B, Li Z, Shen J. A DUF21 domain-containing protein regulates plant dwarfing in watermelon. PLANT PHYSIOLOGY 2024; 196:3091-3104. [PMID: 39268875 DOI: 10.1093/plphys/kiae486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 09/15/2024]
Abstract
Dwarf or semidwarf plant structures are well suited for intensive farming, maximizing yield, and minimizing labor costs. Watermelon (Citrullus lanatus) is classified as an annual vine plant with elongated internodes, yet the mechanism governing watermelon dwarfing remains unclear. In this study, a compact watermelon mutant dwarf, induced by the insertion of transferred DNA (T-DNA), was discovered. Through resequencing, a gene named domain of unknown function 21 (ClDUF21), located downstream of the T-DNA insertion site, was identified as the candidate gene for the dwarf mutant, and its functionality was subsequently confirmed. Watermelon mutants generated through CRISPR/Cas9-mediated knockout of ClDUF21 revealed that homozygous mutants displayed a pronounced dwarfing phenotype, and protein-protein interaction analysis confirmed the direct interaction between ClDUF21 and ClDWF1. Subsequently, we employed CRISPR/Cas9 technology to precisely modify the homologous gene CsDUF21 in cucumber (Cucumis sativus) and performed protein interaction validation between CsDUF21 and CsDWF1, thereby demonstrating that the CsDUF21 gene also exhibits analogous functionality in plant dwarfing. These findings demonstrate that ClDUF21 governs plant dwarfism by modulating the brassinosteroid synthesis pathway via ClDWF1.
Collapse
Affiliation(s)
- Piaoyun Sun
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
- Guangdong Key Laboratory for New Technology Research of Vegetables, Vegetable Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510640, China
| | - Hongjiao Zhao
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Lihong Cao
- College of Horticulture, Shanxi Agricultural University, Taigu 030801, China
| | - Tian Zhang
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Helong Zhang
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Tongwen Yang
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Bosi Zhao
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yanxin Jiang
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Junyang Dong
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Tianrui Chen
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Biao Jiang
- Guangdong Key Laboratory for New Technology Research of Vegetables, Vegetable Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510640, China
| | - Zheng Li
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Junjun Shen
- College of Horticulture, Northwest A&F University, Yangling 712100, Shaanxi, China
| |
Collapse
|
2
|
Gao J, Gerstein M. Representing core gene expression activity relationships using the latent structure implicit in Bayesian networks. Bioinformatics 2024; 40:btae463. [PMID: 39051682 PMCID: PMC11316617 DOI: 10.1093/bioinformatics/btae463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 05/31/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024] Open
Abstract
MOTIVATION Many types of networks, such as co-expression or ChIP-seq-based gene-regulatory networks, provide useful information for biomedical studies. However, they are often too full of connections and difficult to interpret, forming "indecipherable hairballs." RESULTS To address this issue, we propose that a Bayesian network can summarize the core relationships between gene expression activities. This network, which we call the LatentDAG, is substantially simpler than conventional co-expression network and ChIP-seq networks (by two orders of magnitude). It provides clearer clusters, without extraneous cross-cluster connections, and clear separators between modules. Moreover, one can find a number of clear examples showing how it bridges the connection between steps in the transcriptional regulatory network and other networks (e.g. RNA-binding protein). In conjunction with a graph neural network, the LatentDAG works better than other biological networks in a variety of tasks, including prediction of gene conservation and clustering genes. AVAILABILITY AND IMPLEMENTATION Code is available at https://github.com/gersteinlab/LatentDAG.
Collapse
Affiliation(s)
- Jiahao Gao
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, United States
| | - Mark Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, United States
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, United States
- Department of Statistics and Data Science, Yale University, New Haven, CT 06520, United States
- Department of Computer Science, Yale University, New Haven, CT 06520, United States
| |
Collapse
|
3
|
Peng J, Zhang S, Han F, Wang Z. C1QBP is a critical component in the immune response of large yellow croaker (Larimichthys crocea) against visceral white spot disease caused by Pseudomonas plecoglossicida. FISH & SHELLFISH IMMUNOLOGY 2024; 146:109372. [PMID: 38218420 DOI: 10.1016/j.fsi.2024.109372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/27/2023] [Accepted: 01/10/2024] [Indexed: 01/15/2024]
Abstract
The large yellow croaker (Larimichthys crocea) stands as a cornerstone of mariculture in China due to its significant value. However, the threat of Pseudomonas plecoglossicida infection looms large, capable of triggering "visceral white spot disease" and subsequently inflicting severe economic ramifications. Through a prior genome-wide association analysis (GWAS) aimed at understanding the resistance of the large yellow croaker to this ailment, a pivotal player emerged: the complement component 1q binding protein, aptly named LcC1qbp. This protein assumes a crucial role in the activation of the complement system. This study delves deeper into the immune response by examining the expression patterns of LcC1QBP when confronted with P. plecoglossicida. The investigation into gene expression patterns reveals LcC1qbp's widespread presence, with its highest transcriptional abundance identified in the kidney tissues. Upon infection by P. plecoglossicida, the up-regulation of LcC1qbp in major immune organs manifests at both the transcriptional and translational levels. In the context of RNA interference, transcriptome analysis of C1qbp in HEK 293T cells uncovers 1327 differentially expressed genes (DEGs), featuring 41 significant immune genes. This includes pivotal components such as C1S and C3 of the complement system, along with IL11, IL12RB2, and Myd88, among others. The outcomes of enrichment analysis spotlight the prevalence of DEGs within key pathways like immune system development, myeloid leukocyte-mediated immunity, MAPK signaling, and other immune-related routes. By unveiling the immune response mechanisms of the large yellow croaker to P. plecoglossicida infection, this study bolsters our understanding. Furthermore, it lays the groundwork for pursuing effective strategies in both preventing and treating "visceral white spot disease" in the large yellow croaker.
Collapse
Affiliation(s)
- Jia Peng
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Fisheries College, Jimei University, Xiamen, 361000, PR China
| | - Sen Zhang
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Fisheries College, Jimei University, Xiamen, 361000, PR China
| | - Fang Han
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Fisheries College, Jimei University, Xiamen, 361000, PR China.
| | - Zhiyong Wang
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Fisheries College, Jimei University, Xiamen, 361000, PR China
| |
Collapse
|
4
|
Lei Y, Li X, Qin D, Zhang Y, Wang Y. gC1qR: A New Target for Cancer Immunotherapy. Front Immunol 2023; 14:1095943. [PMID: 36776869 PMCID: PMC9909189 DOI: 10.3389/fimmu.2023.1095943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/02/2023] [Indexed: 01/27/2023] Open
Abstract
Although breakthroughs in cancer treatment have been achieved, immunotherapy yields only modest benefits in most patients. There is still a gap in clarifying the immune evasiveness and immune-resistance mechanisms. Identifying other candidate targets for cancer immunotherapy is therefore a clear unmet clinical need. The complement system, a pillar of innate immunity, has recently entered the limelight due to its immunoregulatory functions in the tumor microenvironment (TME). In particular, gC1qR, a receptor for globular heads of C1q, serves as a promising new target and has attracted more attention. gC1qR, also named P32/C1qBP/HABP1, is a multifunctional protein that is overexpressed in various cancers and holds prognostic value. It regulates the tumorigenic, progression and metastatic properties of tumor cells through several downstream signaling pathways, including the Wnt/β-catenin, PKC-NF-κB and Akt/PKB pathways. A few preclinical experiments conducted through gC1qR interventions, such as monoclonal antibody, chimeric antigen receptor T-cell (CAR-T) therapy, and tumor vaccination, have shown encouraging results in anticancer activity. The efficacy may rely on the regulatory role on the TME, induction of tumor cells apoptosis and antiangiogenic activity. Nevertheless, the current understanding of the relationship between cancer immunotherapy and gC1qR remains elusive and often contradictory, posing both opportunities and challenges for therapeutic translation in the clinic. In this review, we focus on the current understanding of gC1qR function in cancer immunology and highlight the vital roles in regulating the TME. We also examines the rationale behind targeting gC1qR and discusses the potential for translating into clinical practice.
Collapse
Affiliation(s)
- Yanna Lei
- Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoyu Li
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China.,Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Diyuan Qin
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China.,Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yugu Zhang
- Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Yongsheng Wang
- Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Desi N, Tong QY, Teh V, Chan JJ, Zhang B, Tabatabaeian H, Tan HQ, Kapeli K, Jin W, Lim CY, Kwok ZH, Tan HT, Wang S, Siew BE, Lee KC, Chong CS, Tan KK, Yang H, Kappei D, Yeo GW, Chung MCM, Tay Y. Global analysis of RNA-binding proteins identifies a positive feedback loop between LARP1 and MYC that promotes tumorigenesis. Cell Mol Life Sci 2022; 79:147. [PMID: 35195778 PMCID: PMC11072786 DOI: 10.1007/s00018-021-04093-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 11/03/2022]
Abstract
In addition to genomic alterations, aberrant changes in post-transcriptional regulation can modify gene function and drive cancer development. RNA-binding proteins (RBPs) are a large class of post-transcriptional regulators that have been increasingly implicated in carcinogenesis. By integrating multi-omics data, we identify LARP1 as one of the most upregulated RBPs in colorectal cancer (CRC) and demonstrate its oncogenic properties. We perform LARP1:RNA interactome profiling and unveil a previously unexplored role for LARP1 in targeting the 3'UTR of oncogenes in CRC. Notably, we identify the proto-oncogenic transcription factor MYC as a key LARP1-regulated target. Our data show that LARP1 positively modulates MYC expression by associating with its 3'UTR. In addition, antisense oligonucleotide-mediated blocking of the interaction between LARP1 and the MYC 3'UTR reduces MYC expression and in vitro CRC growth. Furthermore, a systematic analysis of LARP1:protein interactions reveals IGF2BP3 and YBX1 as LARP1-interacting proteins that also regulate MYC expression and CRC development. Finally, we demonstrate that MYC reciprocally modulates LARP1 expression by targeting its enhancer. In summary, our data reveal a critical, previously uncharacterized role of LARP1 in promoting CRC tumorigenesis, validate its direct regulation of the proto-oncogene MYC and delineate a model of the positive feedback loop between MYC and LARP1 that promotes CRC growth and development.
Collapse
Affiliation(s)
- Ng Desi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Qing Yun Tong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Velda Teh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Jia Jia Chan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Bin Zhang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Hossein Tabatabaeian
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Hui Qing Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Katannya Kapeli
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Wenhao Jin
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Chun You Lim
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Zhi Hao Kwok
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
- Division of Pulmonary and Critical Care, Boston University, Boston, MA, 02118, USA
| | - Hwee Tong Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Shi Wang
- Department of Pathology, National University Health System, Singapore, Singapore
| | - Bei-En Siew
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kuok-Chung Lee
- Division of Colorectal Surgery, University Surgical Cluster, National University Health System, Singapore, Singapore
| | - Choon-Seng Chong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Colorectal Surgery, University Surgical Cluster, National University Health System, Singapore, Singapore
| | - Ker-Kan Tan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Colorectal Surgery, University Surgical Cluster, National University Health System, Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Gene W Yeo
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Cellular and Molecular Medicine, Stem Cell Program and Institute for Genomic Medicine, University of California, La Jolla, San Diego, USA
| | - Maxey Ching Ming Chung
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Yvonne Tay
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
| |
Collapse
|
6
|
Cui Q, Wang C, Liu S, Du R, Tian S, Chen R, Geng H, Subramanian S, Niu Y, Wang Y, Yue D. YBX1 knockdown induces renal cell carcinoma cell apoptosis via Kindlin-2. Cell Cycle 2021; 20:2413-2427. [PMID: 34709966 DOI: 10.1080/15384101.2021.1985771] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Among urological tumors, renal cell carcinoma (RCC) is the third-highest mortality rate tumor, and 20%-30% of RCC patients present with metastases at the time of diagnosis. While the treatment of RCC has been improved over the last few years, its mortality stays high. Y-box binding protein 1 (YBX1) is a well-known oncoprotein that has tumor-promoting functions. YBX1 is widely considered to be an attractive therapeutic target in cancer. To develop novel therapeutics to target YBX1, it is of great importance to understand how YBX1 is finely regulated in cancer. Our previous studies showed that YBX1 in RCC cells significantly promoted cell adhesion, migration, and invasion. However, the role of YBX1 in RCC cells apoptosis has not been reported. In this study, we investigated the effect of YBX1 on cell apoptosis and elucidated the mechanisms involved. Results showed that YBX1 regulated RCC cells apoptosis and reactive oxygen species (ROS) generation via Kindlin-2. These findings indicated that YBX1 inhibited RCC cells apoptosis and may serve as a candidate RCC prognostic marker and a potential therapeutic target. Abbreviations: RCC: Renal cell carcinoma; YBX1: Y-box binding protein 1; ROS: Reactive oxygen species; ccRCC: Clear cell renal cell carcinoma; mccRCC: Metastatic clear cell renal cell carcinoma; G3BP1: Ras-GTPase activating protein SH3 domain-binding proteins 1; SPP1: Secreted phosphoprotein 1; NF-κB: Nuclear factor kappa beta; ECM: Extracellular matrix; EMT: Epithelial-mesenchymal transition; PYCR1: Pyrroline-5-carboxylate reductase 1; MEM: Eagle's Minimum Essential Medium; DMEM: Dulbecco's modified Eagle medium; FBS: Fetal bovine serum; PCR: Polymerase chain reaction; shRNA: Short hairpin RNA; siRNA: Small interfering RNA; BSA: Bovine serum albumin; DCFH-DA: 2,7-Dichlorodihydrofluorescein diacetate; FITC: Fluorescein isothiocyanate; PI: Propidium iodide.
Collapse
Affiliation(s)
- Qiqi Cui
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin China
| | - Chao Wang
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin China
| | - Shuang Liu
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin China
| | - Runxuan Du
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin China
| | - Shaoping Tian
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin China
| | - Ruibing Chen
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin China
| | - Hua Geng
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Saravanan Subramanian
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yuanjie Niu
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin China
| | - Yong Wang
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin China
| | - Dan Yue
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin China
| |
Collapse
|
7
|
p32 promotes melanoma progression and metastasis by targeting EMT markers, Akt/PKB pathway, and tumor microenvironment. Cell Death Dis 2021; 12:1012. [PMID: 34711805 PMCID: PMC8553772 DOI: 10.1038/s41419-021-04311-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 09/19/2021] [Accepted: 10/04/2021] [Indexed: 12/22/2022]
Abstract
Melanoma originates from melanin-producing cells called melanocytes. Melanoma poses a great risk because of its rapid ability to spread and invade new organs. Cellular metastasis involves alteration in the gene expression profile and their transformation from epithelial to mesenchymal state. Despite of several advances, metastatic melanoma being a key cause of therapy failure and mortality remains poorly understood. p32 has been found to be involved in various physiological and pathophysiological conditions. However, the role of p32 in melanoma progression and metastasis remains underexplored. Here, we identify the role of p32 in the malignancy of both murine and human melanoma. p32 knockdown leads to reduced cell proliferation, migration, and invasion in murine and human melanoma cells. Furthermore, p32 promotes in vitro tumorigenesis, inducing oncogenes and EMT markers. Mechanistically, we show p32 regulates tumorigenic and metastatic properties through the Akt/PKB signaling pathway in both murine and human melanoma. Furthermore, p32 silencing attenuates melanoma tumor progression and lung metastasis in vivo, modulating the tumor microenvironment by inhibiting the angiogenesis, infiltration of macrophages, and leukocytes in mice. Taken together, our findings identify that p32 drives melanoma progression, metastasis, and regulates the tumor microenvironment. p32 can be a target of a novel therapeutic approach in the regulation of melanoma progression and metastasis.
Collapse
|
8
|
D'Costa NM, Lowerison MR, Raven PA, Tan Z, Roberts ME, Shrestha R, Urban MW, Monjaras-Avila CU, Oo HZ, Hurtado-Coll A, Chavez-Munoz C, So AI. Y-box binding protein-1 is crucial in acquired drug resistance development in metastatic clear-cell renal cell carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:33. [PMID: 32041631 PMCID: PMC7011538 DOI: 10.1186/s13046-020-1527-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 01/10/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Renal cell carcinoma (RCC) is a highly vascular tumor and patients with low risk metastatic RCC of clear-cell histological sub-type (mccRCC) are treated with tyrosine-kinase inhibitors (TKIs), sunitinib, as the first-line of treatment. Unfortunately, TKI resistance eventually develops, and the underlying molecular mechanism is not well understood. METHODS RCC cell-line with metastatic clear-cell histology (Caki-1), and patient samples were analysed to identify the role of Y-box binding protein 1 (YB-1) and ATP-binding cassette sub-family B member 1 (ABCB-1) in acquired sunitinib-resistance development. Caki-1 was conditioned with increasing sunitinib doses to recapitulate acquired resistance development in clinics. Sunitinib-conditioned and wild-type Caki-1 were subjected to cell viability assay, scratch assay, chicken embryo chorioallantoic membrane engraftment and proteomics analysis. Classical biochemical assays like flow cytometry, immunofluorescent staining, immunohistochemical staining, optical coherence tomography imaging, Western Blot and RT-PCR assays were applied to determine the possible mechanism of sunitinib-resistance development and the effect of drug treatments. Publicly available data was also used to determine the role of YB-1 upregulation in ccRCC and the patients' overall survival. RESULTS We demonstrate that YB-1 and ABCB-1 are upregulated in sunitinib-resistant in vitro, ex vivo, in vivo and patient samples compared to the sensitive samples. This provides evidence to a mechanism of acquired sunitinib-resistance development in mccRCC. Furthermore, our results establish that inhibiting ABCB-1 with elacridar, in addition to sunitinib, has a positive impact on reverting sunitinib-resistance development in in vitro, ex vivo and in vivo models. CONCLUSION This work proposes a targeted therapy (elacridar and sunitinib) to re-sensitize sunitinib-resistant mccRCC and, possibly, slow disease progression.
Collapse
Affiliation(s)
- Ninadh M D'Costa
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Level 6, 2775-Laurel St, Vancouver, BC, V5Z 1M9, Canada.,Vancouver Prostate Centre, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada
| | - Matthew R Lowerison
- Department of Urology, Mayo Clinic College of Medicine and Science, Mayo Clinic, Rochester, MN, USA
| | - Peter A Raven
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Level 6, 2775-Laurel St, Vancouver, BC, V5Z 1M9, Canada
| | - Zheng Tan
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Level 6, 2775-Laurel St, Vancouver, BC, V5Z 1M9, Canada
| | - Morgan E Roberts
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Level 6, 2775-Laurel St, Vancouver, BC, V5Z 1M9, Canada.,Vancouver Prostate Centre, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada
| | - Raunak Shrestha
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Level 6, 2775-Laurel St, Vancouver, BC, V5Z 1M9, Canada
| | | | - Cesar U Monjaras-Avila
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Level 6, 2775-Laurel St, Vancouver, BC, V5Z 1M9, Canada.,Vancouver Prostate Centre, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada
| | - Htoo Zarni Oo
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Level 6, 2775-Laurel St, Vancouver, BC, V5Z 1M9, Canada.,Vancouver Prostate Centre, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada
| | - Antonio Hurtado-Coll
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Level 6, 2775-Laurel St, Vancouver, BC, V5Z 1M9, Canada.,Vancouver Prostate Centre, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada
| | - Claudia Chavez-Munoz
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Level 6, 2775-Laurel St, Vancouver, BC, V5Z 1M9, Canada.,Vancouver Prostate Centre, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada
| | - Alan I So
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Level 6, 2775-Laurel St, Vancouver, BC, V5Z 1M9, Canada. .,Vancouver Prostate Centre, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| |
Collapse
|
9
|
Na Rangsee N, Yanatatsaneejit P, Pisitkun T, Somparn P, Jintaridth P, Topanurak S. Host proteome linked to HPV E7-mediated specific gene hypermethylation in cancer pathways. Infect Agent Cancer 2020; 15:7. [PMID: 32025240 PMCID: PMC6998090 DOI: 10.1186/s13027-020-0271-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022] Open
Abstract
Background Human papillomavirus (HPV) infection causes around 90% of cervical cancer cases, and cervical cancer is a leading cause of female mortality worldwide. HPV-derived oncoprotein E7 participates in cervical carcinogenesis by inducing aberrant host DNA methylation. However, the targeting specificity of E7 methylation of host genes is not fully understood but is important in the down-regulation of crucial proteins of the hallmark cancer pathways. In this study, we aim to link E7-driven aberrations in the host proteome to corresponding gene promoter hypermethylation events in the hope of providing novel therapeutic targets and biomarkers to indicate the progression of cervical cancer. Methods HEK293 cells were transfected with pcDNA3.1-E7 plasmid and empty vector and subjected to mass spectrometry-based proteomic analysis. Down-regulated proteins (where relative abundance was determined significant by paired T-test) relevant to cancer pathways were selected as gene candidates for mRNA transcript abundance measurement by qPCR and expression compared with that in SiHa cells (HPV type 16 positive). Methylation Specific PCR was used to determine promoter hypermethylation in genes downregulated in both SiHa and transfected HEK293 cell lines. The FunRich and STRING databases were used for identification of potential regulatory transcription factors and the proteins interacting with transcription factor gene candidates, respectively. Results Approximately 400 proteins totally were identified in proteomics analysis. The transcripts of six genes involved in the host immune response and cell proliferation (PTMS, C1QBP, BCAP31, CDKN2A, ZMYM6 and HIST1H1D) were down-regulated, corresponding to proteomic results. Methylation assays showed four gene promoters (PTMS, C1QBP, BCAP31 and CDKN2A) were hypermethylated with 61, 55.5, 70 and 78% increased methylation, respectively. Those four genes can be regulated by the GA-binding protein alpha chain, specificity protein 1 and ETS-like protein-1 transcription factors, as identified from FunRich database predictions. Conclusions HPV E7 altered the HEK293 proteome, particularly with respect to proteins involved in cell proliferation and host immunity. Down-regulation of these proteins appears to be partly mediated via host DNA methylation. E7 possibly complexes with the transcription factors of its targeting genes and DNMT1, allowing methylation of specific target gene promoters.
Collapse
Affiliation(s)
- Nopphamon Na Rangsee
- 1Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400 Thailand
| | | | - Trairak Pisitkun
- 3Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Poorichaya Somparn
- 3Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand.,4Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Pornrutsami Jintaridth
- 5Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400 Thailand
| | - Supachai Topanurak
- 1Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400 Thailand
| |
Collapse
|
10
|
Wang Y, Su J, Wang Y, Fu D, Ideozu JE, Geng H, Cui Q, Wang C, Chen R, Yu Y, Niu Y, Yue D. The interaction of YBX1 with G3BP1 promotes renal cell carcinoma cell metastasis via YBX1/G3BP1-SPP1- NF-κB signaling axis. J Exp Clin Cancer Res 2019; 38:386. [PMID: 31481087 PMCID: PMC6720408 DOI: 10.1186/s13046-019-1347-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/25/2019] [Indexed: 12/24/2022] Open
Abstract
Background Renal cell carcinoma (RCC) is a deadly urological tumor that remains largely incurable. Our limited understanding of key molecular mechanisms underlying RCC invasion and metastasis has hampered efforts to identify molecular drivers with therapeutic potential. With evidence from our previous study revealing that nuclear overexpression of YBX1 is associated with RCC T stage and metastasis, we investigated the effects of YBX1 in RCC migration, invasion, and adhesion, and then characterized its interaction with RCC-associated proteins G3BP1 and SPP1. Methods Renal cancer cell lines, human embryonic kidney cells, and clinical samples were analyzed to investigate the functional role of YBX1 in RCC metastasis. YBX1 knockdown cells were established via lentiviral infection and subjected to adhesion, transwell migration, and invasion assay. Microarray, immunoprecipitation, dual-luciferase reporter assay, and classical biochemical assays were applied to characterize the mechanism of YBX1 interaction with RCC-associated proteins G3BP1 and SPP1. Results Knockdown of YBX1 in RCC cells dramatically inhibited cell adhesion, migration, and invasion. Mechanistic investigations revealed that YBX1 interaction with G3BP1 upregulated their downstream target SPP1 in vitro and in vivo, which led to an activated NF-κB signaling pathway. Meanwhile, knockdown of SPP1 rescued the YBX1/G3BP1-mediated activation of NF-κB signaling pathway, and RCC cell migration and invasion. We further showed that YBX1 expression was positively correlated with G3BP1 and SPP1 expression levels in clinical RCC samples. Conclusions YBX1 interacts with G3BP1 to promote metastasis of RCC by activating the YBX1/G3BP1–SPP1–NF-κB signaling axis. Electronic supplementary material The online version of this article (10.1186/s13046-019-1347-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yong Wang
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Jing Su
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China.,Department of Laboratory Medicine, Children's Hospital of Hebei Province, Shijiazhuang, 050031, China
| | - Yiting Wang
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Donghe Fu
- Department of Clinical Laboratory, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Justin E Ideozu
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA.,Human Molecular Genetics Program, Stanley Manne Children's Research Institute, Chicago, IL, 60614, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Hua Geng
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA.,Department of Pediatrics, Feinberg School of Medicine at Northwestern University Chicago, Chicago, IL, 60611, USA
| | - Qiqi Cui
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Chao Wang
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Ruibing Chen
- Department of Genetics, School of Basic Medical Sciences, School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Yixi Yu
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Yuanjie Niu
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Dan Yue
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China. .,Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
11
|
Duan Y, Zhang D. Identification of novel prognostic alternative splicing signature in papillary renal cell carcinoma. J Cell Biochem 2019; 121:672-689. [PMID: 31407370 DOI: 10.1002/jcb.29314] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/15/2019] [Indexed: 12/16/2022]
Abstract
Papillary renal cell carcinoma (pRCC) is a heterogeneous disease containing multifocal or solitary tumors with an aggressive phenotype. Increasing evidence has indicated the involvement of aberrant splicing variants in renal cell cancer, while systematic profiling of aberrant alternative splicing (AS) in pRCC was lacking and largely unknown. In the current study, comprehensive profiling of AS events were performed based on the integration of pRCC cohort from the Cancer Genome Atlas database and SpliceSeq software. With rigorous screening and univariate Cox analysis, a total of 2077 prognoses AS events from 1642 parent genes were identified. Then, stepwise least absolute shrinkage and selection operator method-penalized Cox regression analyses with 10-fold cross-validation followed by multivariate Cox regression were used to construct the prognostic AS signatures within each AS type. And a final 21 AS event-based signature was proposed which showed potent prognostic capability in stratifying patients into low- and high-risk subgroups (P < .0001). Furthermore, time-dependent receiver operating characteristics curves confirmed that the final AS signature was effective and robust in predicting overall survival for pRCC patients with the area under the curve above 0.9 from 1 to 5 years. In addition, splicing correlation network was built to uncover the potential regulatory pattern among prognostic splicing factors and candidate AS events. Besides, gene set enrichment analysis revealed the involvement of these candidates AS events in tumor-related pathways including extracellular matrix organization, oxidative phosphorylation, and P53 signaling pathways. Taken together, our results could contribute to elucidating the underlying mechanism of AS in the oncogenesis process and broaden the novel field of prognostic and clinical application of molecule-targeted approaches in pRCC.
Collapse
Affiliation(s)
- Yi Duan
- Department of Clinical Medicine, Clinical Medical College, Shandong University, Jinan, China.,Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Dong Zhang
- Department of Clinical Medicine, Clinical Medical College, Shandong University, Jinan, China.,Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
12
|
Brandt S, Mertens PR. A remedy for kidney disease successfully alters the cold shock protein response during inflammation. Kidney Int 2019; 90:1148-1150. [PMID: 27884304 DOI: 10.1016/j.kint.2016.07.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 12/18/2022]
Abstract
Kidneys undergoing acute inflammatory responses are characterized by cell infiltration and a cytokinergic milieu. The hazard resides in the perpetuation of inflammation and ensuing fibrosis. In this issue of Kidney International, Wang et al.4 identify the cold shock Y-box binding protein-1 as the key orchestrator of cell infiltration in experimental tubulointerstitial nephritis following ureteral obstruction. Intriguingly, a small molecule previously designed to interfere with Y-box binding protein-1 interactions mediates an anti-inflammatory response and halts fibrogenesis.
Collapse
Affiliation(s)
- Sabine Brandt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Germany
| | - Peter R Mertens
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Germany.
| |
Collapse
|
13
|
Barna J, Dimén D, Puska G, Kovács D, Csikós V, Oláh S, Udvari EB, Pál G, Dobolyi Á. Complement component 1q subcomponent binding protein in the brain of the rat. Sci Rep 2019; 9:4597. [PMID: 30872665 PMCID: PMC6418184 DOI: 10.1038/s41598-019-40788-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 02/19/2019] [Indexed: 12/17/2022] Open
Abstract
Complement component 1q subcomponent binding protein (C1qbp) is a multifunctional protein involved in immune response, energy homeostasis of cells as a plasma membrane receptor, and a nuclear, cytoplasmic or mitochondrial protein. Recent reports suggested its neuronal function, too, possibly in axon maintenance, synaptic function, and neuroplasticity. Therefore, we addressed to identify C1qbp in the rat brain using in situ hybridization histochemistry and immunolabelling at light and electron microscopic level. C1qbp has a topographical distribution in the brain established by the same pattern of C1qbp mRNA-expressing and protein-containing neurons with the highest abundance in the cerebral cortex, anterodorsal thalamic nucleus, hypothalamic paraventricular (PVN) and arcuate nuclei, spinal trigeminal nucleus. Double labelling of C1qbp with the neuronal marker NeuN, with the astrocyte marker S100, and the microglia marker Iba1 demonstrated the presence of C1qbp in neurons but not in glial cells in the normal brain, while C1qbp appeared in microglia following their activation induced by focal ischemic lesion. Only restricted neurons expressed C1qbp, for example, in the PVN, magnocellular neurons selectively contained C1qbp. Further double labelling by using the mitochondria marker Idh3a antibody suggested the mitochondrial localization of C1qbp in the brain, confirmed by correlated light and electron microscopy at 3 different brain regions. Post-embedding immunoelectron microscopy also suggested uneven C1qbp content of mitochondria in different brain areas but also heterogeneity within single neurons. These data suggest a specific function of C1qbp in the brain related to mitochondria, such as the regulation of local energy supply in neuronal cells.
Collapse
Affiliation(s)
- János Barna
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Diána Dimén
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Gina Puska
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Dávid Kovács
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Vivien Csikós
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Szilvia Oláh
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Edina B Udvari
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Gabriella Pál
- Hungarian Defence Forces Military Hospital, Budapest, Hungary
| | - Árpád Dobolyi
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
14
|
Cold shock Y-box binding protein-1 acetylation status in monocytes is associated with systemic inflammation and vascular damage. Atherosclerosis 2018; 278:156-165. [PMID: 30278358 DOI: 10.1016/j.atherosclerosis.2018.09.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/11/2018] [Accepted: 09/18/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND AIMS In dialysis patients, vascular morbidities are highly prevalent and linked to leukocyte extravasation, especially of polarized monocytes. Experimental data demonstrate that phenotypic changes in monocytes require Y-box binding protein-1 (YB-1) upregulation. METHODS We determined YB-1 expression in circulating and vessel-invading monocytes from healthy controls and dialysis patients to correlate results with intima plaque formation and systemic inflammation. RESULTS Compared to healthy subjects, dialysis patients have fewer classical and more intermediate and non-classical monocytes. Post-translationally modified YB-1 (lysine 301/304 acetylation) is detected at high levels in the nucleus of adherent and invading CD14+CD68+ monocytes from umbilical cord and atherosclerosis-prone vessels. The content of non-acetylated YB-1 is significantly decreased (p < 0.001), whereas acetylated YB-1 is correspondingly increased (p < 0.001) throughout all monocyte subpopulations, such that the overall content remains unchanged. CONCLUSIONS In dialysis patients the YB-1 acetylation status is higher with prevailing diabetes and intima plaque formation. Pro-inflammatory mediators TNFα, IL-6, uPAR, CCL2, M-CSF, progranulin, ANP, and midkine, as well as anti-inflammatory IL-10 are significantly increased in dialysis patients, emphasizing a systemic inflammatory milieu. Strong positive correlations of monocytic YB-1 content are seen with ANP, IP-10, IL-6, and IL-10 serum levels. This is the first study demonstrating an association of cold shock protein YB-1 expression with inflammation in hemodialysis patients.
Collapse
|
15
|
Murugesan SN, Yadav BS, Maurya PK, Chaudhary A, Singh S, Mani A. Expression and network analysis of YBX1 interactors for identification of new drug targets in lung adenocarcinoma. J Genomics 2018; 6:103-112. [PMID: 29973960 PMCID: PMC6030768 DOI: 10.7150/jgen.20581] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 08/31/2017] [Indexed: 12/27/2022] Open
Abstract
Y-Box Binding protein 1 (YBX-1) is known to be involved in various types of cancers. It's interactors also play major role in various cellular functions. Present work aimed to study the expression profile of the YBX-1 interactors during lung adenocarcinoma (LUAD). The differential expression analysis involved 57 genes from 95 lung adenocarcinoma samples, construction of gene network and topology analysis. A Total of 43 genes were found to be differentially expressed from which 17 genes were found to be down regulated and 26 genes were up-regulated. We observed that Polyadenylate-binding protein 1 (PABPC1), a protein involved in YBX1 translation, is highly correlated with YBX1. The interaction network analysis for a differentially expressed non-coding RNA Growth Arrest Specific 5 (GAS5) suggests that two proteins namely, Growth Arrest Specific 2 (GAS2) and Peripheral myelin protein 22 (PMP22) are potentially involved in LUAD progression. The network analysis and differential expression suggests that Collagen type 1 alpha 2 (COL1A2) can be potential biomarker and target for LUAD.
Collapse
Affiliation(s)
| | - Birendra Singh Yadav
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, India-211004
| | - Pramod Kumar Maurya
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, India-211004
| | - Amit Chaudhary
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, India-211004
| | - Swati Singh
- Center of Bioinformatics, University of Allahabad, India-211002
| | - Ashutosh Mani
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, India-211004
| |
Collapse
|
16
|
Wang Y, Fu D, Chen Y, Su J, Wang Y, Li X, Zhai W, Niu Y, Yue D, Geng H. G3BP1 promotes tumor progression and metastasis through IL-6/G3BP1/STAT3 signaling axis in renal cell carcinomas. Cell Death Dis 2018; 9:501. [PMID: 29717134 PMCID: PMC5931548 DOI: 10.1038/s41419-018-0504-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/18/2018] [Accepted: 03/20/2018] [Indexed: 12/12/2022]
Abstract
The chronic inflammatory microenvironment within or surrounding the primary renal cell carcinoma (RCC) site promotes oncogenic transformation as well as contributes to the development of metastasis. G3BP stress granule assembly factor 1 (G3BP1) was found to be involved in the regulation of multiple cellular functions. However, its functions in RCC have not been previously explored. Here, we first showed that the expression of G3BP1 is elevated in human RCC and correlates with RCC progression. In cultured RCC cells, knockdown of G3BP1 results in inhibition of tumor cell proliferation, migration, and invasion, consistently with the alteration of epithelial–mesenchymal transition (EMT) and cell proliferative markers, including Cadherins, Vimentin, Snail, Slug, c-Myc, and cyclin D1. Remarkably, knockdown of G3BP1 dramatically impaired the signaling connection of pro-inflammatory cytokine IL-6 stimulation and downstream STAT3 activation in RCC, thus eventually contributing to the disruption of IL-6-elicited RCC migration and metastasis. In addition, in vivo orthotopic tumor xenografts results confirmed that knockdown of G3BP1 suppressed RCC tumor growth and metastasis in mice. Collectively, our findings support the notion that G3BP1 promotes tumor progression and metastasis through IL-6/G3BP1/STAT3 signaling axis in RCC.
Collapse
Affiliation(s)
- Yong Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, 300211, China
| | - Donghe Fu
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China
| | - Yajing Chen
- Research Center of Molecular Biology, Inner Mongolia Medical University, Hohhot, 010059, China
| | - Jing Su
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yiting Wang
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xin Li
- Department of Pharmacology, Tianjin Medical University, Tianjin, 300070, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yuanjie Niu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, 300211, China
| | - Dan Yue
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China.
| | - Hua Geng
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
17
|
Matsumoto K, Kose S, Kuwahara I, Yoshimura M, Imamoto N, Yoshida M. Y-box protein-associated acidic protein (YBAP1/C1QBP) affects the localization and cytoplasmic functions of YB-1. Sci Rep 2018; 8:6198. [PMID: 29670170 PMCID: PMC5906478 DOI: 10.1038/s41598-018-24401-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/26/2018] [Indexed: 01/07/2023] Open
Abstract
The Y-box proteins are multifunctional nucleic acid-binding proteins involved in various aspects of gene regulation. The founding member of the Y-box protein family, YB-1, functions as a transcription factor as well as a principal component of messenger ribonucleoprotein particles (mRNPs) in somatic cells. The nuclear level of YB-1 is well correlated with poor prognosis in many human cancers. Previously, we showed that a Y-box protein–associated acidic protein, YBAP1, which is identical to complement component 1, q subcomponent-binding protein (C1QBP, also called gC1qR, hyaluronan-binding protein 1 [HABP1] or ASF/SF2-associated protein p32), relieves translational repression by YB-1. Here we show that the nuclear localization of YB-1 harboring a point mutation in the cold shock domain was inhibited when co-expressed with YBAP1, whereas cytoplasmic accumulation of the wild-type YB-1 was not affected. We showed that YBAP1 inhibited the interaction between YB-1 and transportin 1. In the cytoplasm, YBAP1 affected the accumulation of YB-1 to processing bodies (P-bodies) and partially abrogated the mRNA stabilization by YB-1. Our results, indicating that YBAP1/C1QBP regulates the nucleo-cytoplasmic distribution of YB-1 and its cytoplasmic functions, are consistent with a model that YBAP1/C1QBP acts as an mRNP remodeling factor.
Collapse
Affiliation(s)
- Ken Matsumoto
- Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, RIKEN, Wako, Saitama, Japan. .,PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan. .,Molecular Entomology Laboratory, RIKEN, Wako, Saitama, Japan.
| | - Shingo Kose
- Cellular Dynamics Laboratory, RIKEN Cluster for Pioneering Research (CPR), RIKEN, Wako, Saitama, Japan
| | - Iku Kuwahara
- Molecular Entomology Laboratory, RIKEN, Wako, Saitama, Japan
| | - Mami Yoshimura
- Molecular Entomology Laboratory, RIKEN, Wako, Saitama, Japan
| | - Naoko Imamoto
- Cellular Dynamics Laboratory, RIKEN Cluster for Pioneering Research (CPR), RIKEN, Wako, Saitama, Japan
| | - Minoru Yoshida
- Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, RIKEN, Wako, Saitama, Japan
| |
Collapse
|
18
|
Hohlfeld R, Brandt S, Bernhardt A, Gorny X, Schindele D, Jandrig B, Schostak M, Isermann B, Lindquist JA, Mertens PR. Crosstalk between Akt signaling and cold shock proteins in mediating invasive cell phenotypes. Oncotarget 2018; 9:19039-19049. [PMID: 29721182 PMCID: PMC5922376 DOI: 10.18632/oncotarget.24886] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/25/2018] [Indexed: 12/31/2022] Open
Abstract
Cold shock proteins are up-regulated by cellular stress and orchestrate inflammatory responses, cell proliferation, and differentiation. Enhanced cold shock protein expression promotes malignant cell transformation; up-regulation is detected in most cancers and associated with poor prognosis. Akt1, a serine/threonine kinase, is a potent oncogene, which activates pro-proliferative and anti-apoptotic signaling pathways, and phosphorylates the cold shock domain. Unexpectedly, chicken-YB-1 abrogates PI3K-Akt-dependent oncogenic cell transformation in embryonic fibroblasts. Here, we addressed the question whether chicken and human Y-box binding protein-1 (YB-1) act differently on cell transformation, and how a related protein, DNA-binding protein-A (DbpA) behaves in comparison. NIH3T3 cells were transduced with lentiviral vectors encoding for myristoylated (constitutive active) Akt1, YB-1, DbpA, or shRNA targeting YB-1 expression. Colony formation assays showed that human YB-1 acts similar to chicken on Akt-dependent cell transformation. This activity was not titratable. Given the correlation of nuclear YB-1 and upregulated DbpA expression in a series of clear cell renal cell carcinomas (n = 40) the colony formation assay was extended to include ectopic DbpA expression. DbpA alone prominently induced cell transformation, which was enhanced when constitutive active Akt1 or concomitant YB-1 expression was present. Notably, co-expression of DbpA together with YB-1 abrogated the repressive effect on Akt1 signaling observed with YB-1 alone. Macroscopically, some colonies yielded a remarkable “invasive” phenotype. Thus, cold shock proteins may convey profound anti- and pro-oncogenic effects on Akt-dependent cell transformation. DbpA is able to overcome the anti-oncogenic effects seen with combined YB-1 and Akt signaling in an in vitro model of colonial growth.
Collapse
Affiliation(s)
- Raphael Hohlfeld
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Sabine Brandt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anja Bernhardt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Xenia Gorny
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Daniel Schindele
- Clinic of Urology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Burkhard Jandrig
- Clinic of Urology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Martin Schostak
- Clinic of Urology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jonathan A Lindquist
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Peter R Mertens
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
19
|
Wang Y, Su J, Fu D, Wang Y, Chen Y, Chen R, Qin G, Zuo J, Yue D. The Role of YB1 in Renal Cell Carcinoma Cell Adhesion. Int J Med Sci 2018; 15:1304-1311. [PMID: 30275756 PMCID: PMC6158664 DOI: 10.7150/ijms.25580] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/28/2018] [Indexed: 11/05/2022] Open
Abstract
Background: Y-box binding protein 1 (YB1) is a multifunctional protein involved in many processes related to cancer progression and metastasis. Methods: In this study, we constructed YB1 knockdown stable renal cell carcinoma (RCC) cell line 786-0. The gene expression profile of 786-0 was performed by DNA microarray analysis to identify genes that were regulated by YB1. Real-time PCR and western blotting were used to test the genes and proteins expression. Transforming growth factor-β (TGF-β) activity was detected by dual-luciferase reporter assay. Cell adhesion assay was used to determine RCC cell adhesion ability. Results: Pathway analysis revealed that YB1 knockdown influenced cell adhesion molecules (CAMs). We further verified four genes (CLDN4, NRXN3, ITGB8, and VCAN) related to CAMs by real-time PCR, and confirmed that YB1 regulated the expression of ITGB8 in RCC. Functional assays demonstrated that knockdown of YB1 significantly inhibited the cell adhesion of 786-0 cells in vitro. In addition, YB1 affected TGF-β activation. Conclusion: Our study demonstrated that YB1 modulated the adhesion ability of renal cell carcinoma cells by regulating ITGB8 and TGF-β.
Collapse
Affiliation(s)
- Yong Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China
| | - Jing Su
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China
| | - Donghe Fu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China.,Department of Clinical Laboratory, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Yiting Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China
| | - Yajing Chen
- Research Center of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
| | - Ruibing Chen
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Guoxuan Qin
- School of Microelectronics, Tianjin University, Tianjin 300072, China
| | - Jing Zuo
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China
| | - Dan Yue
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
20
|
Wang Y, Wang Y, Xu L, Lu X, Fu D, Su J, Geng H, Qin G, Chen R, Quan C, Niu Y, Yue D. CD4 + T cells promote renal cell carcinoma proliferation via modulating YBX1. Exp Cell Res 2017; 363:95-101. [PMID: 29289594 DOI: 10.1016/j.yexcr.2017.12.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/14/2017] [Accepted: 12/27/2017] [Indexed: 12/25/2022]
Abstract
Renal cell carcinoma (RCC) is a common urologic tumor and the third leading cause of death among urological tumors. Recent studies demonstrate that RCC tumors are more heavily infiltrated by lymphocytes than other cancers. However, the exact roles played by CD4 + T cells in RCC proliferation remain unknown. In this study, we cocultured RCC cells with CD4 + T cells. Stable knockdown of YBX1 in RCC cells was constructed. The effects of CD4 + T cells, TGFβ1 and YBX1 on RCC cells were investigated using cell viability assays. In situ RCC nude mouse model was used to observe the tumor growth. The potential mechanisms of CD4 + T cells and YBX1 in RCC cells proliferation were explored by qRT-PCR and western blot. Expression of CD4, Foxp3 and TGFβ1 in RCC were quantified by immunohistochemical staining. The results indicated that CD4, Foxp3 and TGFβ1 were significantly up-regulated in RCC tissues. Human clinical sample and in vitro cell lines studies showed that RCC cells had better capacity than its surrounding normal kidney epithelial cells to recruit the CD4 + T cells. In vivo mouse model studies were consistent with the results by in vitro cell lines studies showing infiltrating T cells enhanced RCC cell proliferation. qRT-PCR and western blot exhibited that CD4 + T cells could enhance RCC cell proliferation via activating YBX1/HIF2α signaling pathway. Furthermore, CD4 + T cells functioned through inducing TGFβ1 expression. In a word, infiltrating CD4 + T cells promoted TGFβ1 expression in both RCC and T cells and regulated RCC cells proliferation via modulating TGFβ1/YBX1/ HIF2α signals.
Collapse
Affiliation(s)
- Yong Wang
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Yiting Wang
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Liang Xu
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Xianqi Lu
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Donghe Fu
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Jing Su
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Hua Geng
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Guoxuan Qin
- School of Microelectronics, Tianjin University, Tianjin 300072, China
| | - Ruibing Chen
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Changyi Quan
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Yuanjie Niu
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China
| | - Dan Yue
- The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, and School of Medical Laboratory, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
21
|
Chen R, Liu Y, Zhuang H, Yang B, Hei K, Xiao M, Hou C, Gao H, Zhang X, Jia C, Li L, Li Y, Zhang N. Quantitative proteomics reveals that long non-coding RNA MALAT1 interacts with DBC1 to regulate p53 acetylation. Nucleic Acids Res 2017; 45:9947-9959. [PMID: 28973437 PMCID: PMC5622371 DOI: 10.1093/nar/gkx600] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 07/05/2017] [Indexed: 12/17/2022] Open
Abstract
Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a broadly expressed lncRNA involved in many aspects of cellular processes. To further delineate the underlying molecular mechanism, we employed a high-throughput strategy to characterize the interacting proteins of MALAT1 by combining RNA pull-down, quantitative proteomics, bioinformatics, and experimental validation. Our approach identified 127 potential MALAT1-interacting proteins and established a highly connected MALAT1 interactome network consisting of 788 connections. Gene ontology annotation and network analysis showed that MALAT1 was highly involved in five biological processes: RNA processing; gene transcription; ribosomal proteins; protein degradation; and metabolism regulation. The interaction between MALAT1 and depleted in breast cancer 1 (DBC1) was validated using RNA pull-down and RNA immunoprecipitation. Further mechanistic studies reveal that MALAT1 binding competes with the interaction between sirtuin1 (SIRT1) and DBC1, which then releases SIRT1 and enhances its deacetylation activity. Consequently, the deacetylation of p53 reduces the transcription of a spectrum of its downstream target genes, promotes cell proliferation and inhibits cell apoptosis. Our results uncover a novel mechanism by which MALAT1 regulates the activity of p53 through the lncRNA–protein interaction.
Collapse
Affiliation(s)
- Ruibing Chen
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
| | - Yun Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
| | - Hao Zhuang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
- Department of Hepatic Biliary Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450000, China
| | - Baicai Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
| | - Kaiwen Hei
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
| | - Mingming Xiao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
| | - Chunyu Hou
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
| | - Huajun Gao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
| | - Xinran Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
| | - Chenxi Jia
- Chemistry Department & School of Pharmacy, University of Wisconsin at Madison, Madison, WI 53705, USA
- National Center for Protein Sciences-Beijing, State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China
| | - Lingjun Li
- Chemistry Department & School of Pharmacy, University of Wisconsin at Madison, Madison, WI 53705, USA
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Yongmei Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
- To whom correspondence should be addressed. Tel: +86 22 83336531; Fax: +86 22 83336560; . Correspondence may also be addressed to Ning Zhang. Tel: +86 22 83336531; Fax: +86 22 83336560;
| | - Ning Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy; Department of Genetics & Department of Medical Microbiology, School of Basic Medical Sciences; Research Center of Basic Medical Sciences; Tianjin Medical University, Tianjin 300070, China
- To whom correspondence should be addressed. Tel: +86 22 83336531; Fax: +86 22 83336560; . Correspondence may also be addressed to Ning Zhang. Tel: +86 22 83336531; Fax: +86 22 83336560;
| |
Collapse
|
22
|
Wang Y, Su J, Yuan B, Fu D, Niu Y, Yue D. The role of C1QBP in CSF-1-dependent PKCζ activation and macrophage migration. Exp Cell Res 2017; 362:11-16. [PMID: 28965866 DOI: 10.1016/j.yexcr.2017.09.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/23/2017] [Accepted: 09/26/2017] [Indexed: 11/30/2022]
Abstract
Macrophages view as double agents in tumor progression. Trafficking of macrophages to the proximity of tumors is mediated by colony-stimulating factor-1 (CSF-1), a growth factor. In this study, we investigated the role of complement1q-binding protein (C1QBP)/ atypical protein kinase C ζ (PKCζ) in CSF-1-induced macrophage migration. Disruption of C1QBP expression impaired chemotaxis and adhesion of macrophage. Phosphorylation of PKCζ is an essential component in macrophage chemotaxis signaling pathway. C1QBP could interact with PKCζ in macrophage. C1QBP knockdown inhibited CSF-1 induced phosphorylation of PKCζ and integrin-β1. However, C1QBP knockdown didn't affect the phosphorylation of PKCζ induced by MCP-1. Furthermore, CSF-1 from RCC cell condition medium promoted macrophage chemotaxis and adhesion. Taken together, our results demonstrated that C1QBP plays an essential role in CSF-1 induced migration of macrophages.
Collapse
Affiliation(s)
- Yong Wang
- Department of Urology, Tianjin Medical University Second Hospital, Tianjin Institute of Urology, Tianjin Medical University, Tianjin 300211, China
| | - Jing Su
- School of Laboratory Medicine, Tianjin Medical University, Tianjin 300203, China
| | - Bo Yuan
- School of Laboratory Medicine, Tianjin Medical University, Tianjin 300203, China
| | - Donghe Fu
- School of Laboratory Medicine, Tianjin Medical University, Tianjin 300203, China
| | - Yuanjie Niu
- Department of Urology, Tianjin Medical University Second Hospital, Tianjin Institute of Urology, Tianjin Medical University, Tianjin 300211, China
| | - Dan Yue
- School of Laboratory Medicine, Tianjin Medical University, Tianjin 300203, China.
| |
Collapse
|
23
|
C1QBP suppresses cell adhesion and metastasis of renal carcinoma cells. Sci Rep 2017; 7:999. [PMID: 28428626 PMCID: PMC5430506 DOI: 10.1038/s41598-017-01084-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/23/2017] [Indexed: 11/09/2022] Open
Abstract
Complement component 1q subcomponent binding protein (C1QBP) is a ubiquitously expressed cellular protein and can be upregulated or activated in a variety of malignant tumors, including those from thyroid, colon and breast, but its role remains unclear in renal cell carcinoma (RCC). In this study, C1QBP knockdown in RCC cell influenced expression of multiple genes associated with cell adhesion, among which L1 cell adhesion molecule (L1CAM) was significantly higher upon a reduction of C1QBP. In turn, cell adhesion and invasion abilities were significantly increased with increased metastasis to lung and liver in vivo. C1QBP may regulate RCC cell adhesion and invasion through influencing the p-GSK3/β-Catenin/L1CAM expression. Over all, our study demonstrated that C1QBP could regulate RCC metastasis by regulating the GSK3/β-Catenin/L1CAM signaling pathway.
Collapse
|
24
|
Yue D, Wang Y, Sun Y, Niu Y, Chang C. C1QBP Regulates YBX1 to Suppress the Androgen Receptor (AR)-Enhanced RCC Cell Invasion. Neoplasia 2017; 19:135-144. [PMID: 28107702 PMCID: PMC5247285 DOI: 10.1016/j.neo.2016.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/26/2016] [Accepted: 12/01/2016] [Indexed: 12/20/2022]
Abstract
Early studies suggested that the androgen receptor (AR) might play important roles to promote the renal cell carcinoma (RCC) progression; however, the detailed mechanisms remain unclear. Here we demonstrated the higher YBX1 expression with lower C1QBP expression in human RCC clinical tissues, and the intensity of C1QBP was negatively correlated with the YBX1 nuclear expression. Mechanism dissection found C1QBP could interact with YBX1 to suppress the YBX1 activation via altering the YBX1 phosphorylation and nuclear translocation in RCC cells. The consequences of such suppression of YBX1 might then result in suppressing the RCC cell migration and invasion that involved altering the AR-modulated MMP9 signals. Interruption of this newly identified C1QBP→YBX1→AR→MMP9-suppressed RCC cell invasion pathway via targeting YBX1 or AR partially reversed the RCC cell invasion. Importantly, results from in vivo mouse model with orthotopic implantation of RCC OSRC2 cells into the left renal capsule also confirmed in vitro cell line studies showing targeting YBX1 could suppress RCC cell invasion via regulation of AR/MMP9 signals. Collectively, these data suggest that C1QBP could regulate YBX1 to suppress the AR-enhanced RCC cell invasion. Targeting this newly identified C1QBP/YBX1/AR/MMP9 signal pathway may provide a new potential therapy to better suppress RCC metastasis.
Collapse
Affiliation(s)
- Dan Yue
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology and School of Laboratory Medicine, Tianjin Medical University, Tainjin 300203, China; George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Yong Wang
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology and School of Laboratory Medicine, Tianjin Medical University, Tainjin 300203, China; George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Yin Sun
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Yuanjie Niu
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology and School of Laboratory Medicine, Tianjin Medical University, Tainjin 300203, China.
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA; Sex Hormone Research Center, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
25
|
Chinello C, L'imperio V, Stella M, Smith AJ, Bovo G, Grasso A, Grasso M, Raimondo F, Pitto M, Pagni F, Magni F. The proteomic landscape of renal tumors. Expert Rev Proteomics 2016; 13:1103-1120. [PMID: 27748142 DOI: 10.1080/14789450.2016.1248415] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Renal cell carcinoma (RCC) is the most fatal of the common urologic cancers, with approximately 35% of patients dying within 5 years following diagnosis. Therefore, there is a need for non-invasive markers that are capable of detecting and determining the severity of small renal masses at an early stage in order to tailor treatment and follow-up. Proteomic studies have proved to be very useful in the study of tumors. Areas covered: In this review, we will detail the current knowledge obtained by the different proteomic approaches, focusing on MS-based strategies, used to investigate RCC biology in order to identify diagnostic, prognostic and predictive biomarkers on tissue, cultured cells and biological fluids. Expert commentary: Currently, no reliable biomarkers or targets for RCC have been translated into the clinical setting. Moreover, despite the efforts of proteomics and other -omics disciplines, only a small number of them have been observed as shared targets between the different analytical platforms and biological specimens. The difficulty to define a specific molecular pattern for RCC and its subtypes highlights a peculiar profile and a heterogeneity that must be taken into account in future studies.
Collapse
Affiliation(s)
- Clizia Chinello
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Vincenzo L'imperio
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Martina Stella
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Andrew James Smith
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Giorgio Bovo
- b Pathology unit , San Gerardo Hospital , Monza , Italy
| | - Angelica Grasso
- c Department of Specialistic Surgical Sciences, Urology unit , Ospedale Maggiore Policlinico Foundation , Milano , Italy
| | - Marco Grasso
- d Department of Urology , San Gerardo Hospital , Monza , Italy
| | - Francesca Raimondo
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Marina Pitto
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Fabio Pagni
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Fulvio Magni
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| |
Collapse
|
26
|
Liu Q, Tao T, Liu F, Ni R, Lu C, Shen A. Hyper-O-GlcNAcylation of YB-1 affects Ser102 phosphorylation and promotes cell proliferation in hepatocellular carcinoma. Exp Cell Res 2016; 349:230-238. [PMID: 27751836 DOI: 10.1016/j.yexcr.2016.10.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 10/09/2016] [Accepted: 10/13/2016] [Indexed: 11/16/2022]
Abstract
As an essential post-translational modification, O-GlcNAcylation has been thought to be able to modulate various nuclear and cytoplasmic proteins and is emerging as a key regulator of multiple biological processes, such as transcription, cell growth, signal transduction, and cell motility. Recently, authoritative glycomics analyses have reported extensive crosstalk between O-GlcNAcylation and phosphorylation, which always dynamically interplay with each other and regulate signaling, transcription, and other cellular processes. Also, plentiful studies have shown close correlation between YB-1 phosphorylation and tumorigenesis. Therefore, our study aimed to determine whether YB-1 was O-GlcNAc modified and whether such modification could interact with its phosphorylation during the process of HCC development. Western blot and immunohistochemistry were firstly conducted to reveal obvious up-regulation of YB-1, OGT and O-GlcNAc modification in HCC tissues. What is more, not only YB-1 was identified to be O-GlcNAcylated but hyper-O-GlcNAcylation was demonstrated to facilitate HCC cell proliferation in a YB-1 dependent manner. Moreover, we detected four specific O-GlcNAc sites and confirmed T126A to be the most effective mutant in HCC cell proliferation via close O-GlcNAcylation-phosphorylation interaction. Even more interestingly, we discovered that T126A-induced HCC cell retardation and subdued transcriptional activity of YB-1 could be partially reversed by T126A/S102E mutant. From all above, it is not difficult to find that glycosylated-YB-1 mainly enhanced cell proliferation through congenerous actions with YB-1 phosphorylation and thus played indispensable roles in fine-tuning cell proliferation and procession of HCC.
Collapse
Affiliation(s)
- Qingqing Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 19 Qi-xiu Road, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Tao Tao
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 19 Qi-xiu Road, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Fang Liu
- Key Laboratory of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Runzhou Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China
| | - Cuihua Lu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China.
| | - Aiguo Shen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 19 Qi-xiu Road, Nantong 226001, Jiangsu Province, People's Republic of China; Key Laboratory of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China.
| |
Collapse
|
27
|
Chen R, Xiao M, Gao H, Chen Y, Li Y, Liu Y, Zhang N. Identification of a novel mitochondrial interacting protein of C1QBP using subcellular fractionation coupled with CoIP-MS. Anal Bioanal Chem 2016; 408:1557-64. [DOI: 10.1007/s00216-015-9228-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/03/2015] [Accepted: 11/27/2015] [Indexed: 12/22/2022]
|