1
|
Lucca MS, Bustamante-Filho IC, Ulguim RR, Gianluppi RDF, Evaristo JAM, Nogueira FCS, Timmers LFSM, Mellagi APG, Wentz I, Bortolozzo FP. Proteomic analysis of boar seminal plasma: Putative markers for fertility based on capacity of semen preservation at 17°C. Mol Reprod Dev 2024; 91:e23735. [PMID: 38282317 DOI: 10.1002/mrd.23735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/20/2023] [Accepted: 01/13/2024] [Indexed: 01/30/2024]
Abstract
Boar seminal plasma (SP) proteins were associated with differences on sperm resistance to cooling at 17°C. However, information about seminal plasma proteins in boars classified by capacity of semen preservation and in vivo fertility remains lacking. Thus, the objective was to evaluate the SP proteome in boars classified by capacity of semen preservation and putative biomarkers for fertility. The ejaculates from high-preservation (HP) showed higher progressive motility during all 5 days than the low-preservation (LP) boars. There was no difference for farrowing rate between ejaculates from LP (89.7%) and HP boars (88.4%). The LP boars presented lower total piglets born (14.0 ± 0.2) than HP (14.8 ± 0.2; p < 0.01). A total of 257 proteins were identified, where 184 were present in both classes of boar, and 41 and 32 were identified only in LP and HP boars, respectively. Nine proteins were differently expressed: five were more abundant in HP (SPMI, ZPBP1, FN1, HPX, and C3) and four in LP boars (B2M, COL1A1, NKX3-2, and MPZL1). The HP boars had an increased abundance of SP proteins related to sperm resistance and fecundation process which explains the better TPB. LP boars had a higher abundance of SP proteins associated with impaired spermatogenesis.
Collapse
Affiliation(s)
- Matheus S Lucca
- Departamento de Medicina Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul-UFRGS, Setor de Suínos, Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Rafael R Ulguim
- Departamento de Medicina Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul-UFRGS, Setor de Suínos, Porto Alegre, Rio Grande do Sul, Brazil
| | - Rafael D F Gianluppi
- Departamento de Medicina Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul-UFRGS, Setor de Suínos, Porto Alegre, Rio Grande do Sul, Brazil
| | - Joseph A M Evaristo
- Laboratory of Proteomics, LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio C S Nogueira
- Laboratory of Proteomics, LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Proteomics Unit, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luís F S M Timmers
- Laboratório de Biotecnologia, Universidade do Vale do Taquari-Univates, Lajeado, Brazil
| | - Ana P G Mellagi
- Departamento de Medicina Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul-UFRGS, Setor de Suínos, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ivo Wentz
- Departamento de Medicina Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul-UFRGS, Setor de Suínos, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernando P Bortolozzo
- Departamento de Medicina Animal, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul-UFRGS, Setor de Suínos, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
2
|
Juan-Guadarrama DG, Beltrán-Navarro YM, Reyes-Cruz G, Vázquez-Prado J. Ephexin3/ARHGEF5 Together with Cell Migration Signaling Partners within the Tumor Microenvironment Define Prognostic Transcriptional Signatures in Multiple Cancer Types. Int J Mol Sci 2023; 24:16427. [PMID: 38003617 PMCID: PMC10671824 DOI: 10.3390/ijms242216427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer cell migration involves a repertoire of signaling proteins that lead cytoskeleton reorganization as a critical step in metastatic dissemination. RhoGEFs are multidomain effectors that integrate signaling inputs to activate the molecular switches that orchestrate actin cytoskeleton reorganization. Ephexins, a group of five RhoGEFs, play oncogenic roles in invasive and metastatic cancer, leading to a mechanistic hypothesis about their function as signaling nodes assembling functional complexes that guide cancer cell migration. To identify clinically significant Ephexin signaling partners, we applied three systematic data mining strategies, based on the screening of essential Ephexins in multiple cancer cell lines and the identification of coexpressed signaling partners in the TCGA cancer patient datasets. Based on the domain architecture of encoded proteins and gene ontology criteria, we selected Ephexin signaling partners with a role in cytoskeletal reorganization and cell migration. We focused on Ephexin3/ARHGEF5, identified as an essential gene in multiple cancer cell types. Based on significant coexpression data and coessentiality, the signaling repertoire that accompanies Ephexin3 corresponded to three groups: pan-cancer, cancer-specific and coessential. To further select the Ephexin3 signaling partners likely to be relevant in clinical settings, we first identified those whose high expression was statistical linked to shorter patient survival. The resulting Ephexin3 transcriptional signatures represent significant accumulated risk, predictive of shorter survival, in 17 cancer types, including PAAD, LUAD, LGG, OSC, AML, KIRC, THYM, BLCA, LIHC and UCEC. The signaling landscape that accompanies Ephexin3 in various cancer types included the tyrosine kinase receptor MET and the tyrosine phosphatase receptor PTPRF, the serine/threonine kinases MARK2 and PAK6, the Rho GTPases RHOD, RHOF and RAC1, and the cytoskeletal regulator DIAHP1. Our findings set the basis to further explore the role of Ephexin3/ARHGEF5 as an essential effector and signaling hub in cancer cell migration.
Collapse
Affiliation(s)
- Dante Gustavo Juan-Guadarrama
- Department of Pharmacology, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Yarely Mabell Beltrán-Navarro
- Department of Pharmacology, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Guadalupe Reyes-Cruz
- Department of Cell Biology, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico
| | - José Vázquez-Prado
- Department of Pharmacology, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico
| |
Collapse
|
3
|
Hasegawa N, Hongo M, Okada M, Kuga T, Abe Y, Adachi J, Tomonaga T, Yamaguchi N, Nakayama Y. Phosphotyrosine proteomics in cells synchronized at monopolar cytokinesis reveals EphA2 as functioning in cytokinesis. Exp Cell Res 2023; 432:113783. [PMID: 37726045 DOI: 10.1016/j.yexcr.2023.113783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
Cytokinesis is the final step of the cell division in which cellular components are separated into two daughter cells. This process is regulated through the phosphorylation of different classes of proteins by serine/threonine (Ser/Thr) kinases such as Aurora B and Polo-like kinase 1 (PLK1). Conversely, the role of phosphorylation at tyrosine residues during cytokinesis has not been studied in detail yet. In this study, we performed a phosphotyrosine proteomic analysis of cells undergoing monopolar cytokinesis synchronized by using the Eg5 inhibitor (+)-S-trityl-l-cysteine (STLC) and the CDK1 inhibitor RO-3306. Phosphotyrosine proteomics gave 362 tyrosine-phosphorylated peptides. Western blot analysis of proteins revealed tyrosine phosphorylation in mitogen-activated protein kinase 14 (MAPK14), vimentin, ephrin type-A receptor 2 (EphA2), and myelin protein zero-like protein 1 (MPZL1) during monopolar cytokinesis. Additionally, we demonstrated that EphA2, a protein with unknown function during cytokinesis, is involved in cytokinesis. EphA2 knockdown accelerated epithelial cell transforming 2 (Ect2) knockdown-induced multinucleation, suggesting that EphA2 plays a role in cytokinesis in a particular situation. The list also included many proteins previously reported to play roles during cytokinesis. These results evidence that the identified phosphopeptides facilitate the identification of novel tyrosine phosphorylation signaling involved in regulating cytokinesis.
Collapse
Affiliation(s)
- Nanami Hasegawa
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Mayue Hongo
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Misaki Okada
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Takahisa Kuga
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan; Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, Osaka 573-0101, Japan
| | - Yuichi Abe
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; Division of Molecular Diagnostics, Aichi Cancer Center, Nagoya 464-8681, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; Proteobiologics Co., Ltd., Osaka 567-0085, Japan
| | - Naoto Yamaguchi
- Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Yuji Nakayama
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| |
Collapse
|
4
|
Ge Q, Zhou C, Zang C, Li C, Hong H, Wang K, Chen L, Zhu H, Wang A. MPZL1 suppresses the cancer stem-like properties of lung cancer through β-catenin/TCF4 signaling. Funct Integr Genomics 2023; 23:304. [PMID: 37726580 DOI: 10.1007/s10142-023-01232-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023]
Abstract
This study was designed to explore the influence of myelin protein zero-like protein 1 (MPZL1) on the stem-like properties of cancer cells and the underlying mechanism in lung adenocarcinoma. Real-time quantitative polymerase chain reaction (RT-qPCR) was utilized to evaluate mRNA expression level. CCK8, wound healing, and transwell assays were applied to assess cell proliferation, migration, and invasion. Tumorsphere-formation assay was utilized to assess cancer stem cell-like properties. LF3 was used to block the β-catenin/Transcription factor 4 (TCF-4) signaling. Xenograft nude mouse model was conducted; tumor weight and volume were recorded. Western blot assay was utilized to detect the expression levels of CD44, CD133, β-catenin, TCF-4, and MPZL1. Following MPZL1 knockdown, the mRNA expression levels of MPZL1, β-catenin, and TCF-4 were inhibited, while the mRNA expression levels of the above genes were increased after the MPZL1 overexpression. MPZL1 knockdown suppressed cell proliferation, migration, and invasion, reduced the tumorsphere-formation capacity, and restrained the expression levels of CD44 and CD133. However, MPZL1 overexpression promoted the cell proliferation, migration, and invasion, enhanced the tumorsphere-formation capacity, and increased the expression levels of CD44 and CD133. Interestingly, LF3 treatment partially revised the effect of MPZL1 overexpression. These findings were further corroborated by in vivo experiments. We concluded that MPZL1 could suppress the lung adenocarcinoma cells' proliferation, migration, invasion, and lung cancer stem cells characteristics. The underlying mechanism is involved in the activation of β-catenin/TCF-4 signaling.
Collapse
Affiliation(s)
- Qiao Ge
- Department of Thoracic Surgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Chao Zhou
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chao Zang
- Department of Thoracic Surgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Chao Li
- Department of Thoracic Surgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Haining Hong
- Department of Thoracic Surgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Kangwu Wang
- Department of Thoracic Surgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Liwei Chen
- Department of Thoracic Surgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Haonan Zhu
- Department of Thoracic Surgery, Fu Yang People's Hospital, Fuyang, Anhui, China.
| | - Ansheng Wang
- Department of Thoracic Surgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.
| |
Collapse
|
5
|
Fu Y, Sui Y, Zhao Y, Jiang J, Wang X, Cui J, Fu X, Xing S, Zhao ZJ. PZR promotes tumorigenicity of lung cancer cells by regulating cell migration and invasion via modulating oxidative stress and cell adhesion. Aging (Albany NY) 2023; 15:204771. [PMID: 37279992 PMCID: PMC10292906 DOI: 10.18632/aging.204771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/17/2023] [Indexed: 06/08/2023]
Abstract
PZR is a transmembrane glycoprotein encoded by the MPZL1 gene. It serves as a specific binding protein and substrate of tyrosine phosphatase SHP-2 whose mutations cause developmental diseases and cancers. Bioinformatic analyses of cancer gene databases revealed that PZR is overexpressed in lung cancer and correlated with unfavorable prognosis. To investigate the role of PZR in lung cancer, we employed the CRISPR technique to knockout its expression and recombinant lentiviruses to overexpress it in lung adenocarcinoma SPC-A1 cells. While knockout of PZR reduced colony formation, migration, and invasion, overexpression of PZR had the opposite effects. Furthermore, when implanted in immunodeficient mice, PZR-knockout SPC-A1 cells showed suppressed tumor-forming ability. Finally, the underlying molecular mechanism for these functions of PZR is its positive role in activating tyrosine kinases FAK and c-Src and in maintaining the intracellular level of reactive oxygen species (ROS). In conclusion, our data indicated that PZR plays an important role in lung cancer development, and it may serve as a therapeutic target for anti-cancer development and as a biomarker for cancer prognosis.
Collapse
Affiliation(s)
- Ying Fu
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, China
| | - Yuan Sui
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, China
| | - Yuming Zhao
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, China
| | - Jianzhuo Jiang
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, China
| | - Xueyuan Wang
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, China
| | - Jiarui Cui
- Department of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Xueqi Fu
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, China
| | - Shu Xing
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, China
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
6
|
Smirnova EV, Rakitina TV, Ziganshin RH, Saratov GA, Arapidi GP, Belogurov AA, Kudriaeva AA. Identification of Myelin Basic Protein Proximity Interactome Using TurboID Labeling Proteomics. Cells 2023; 12:cells12060944. [PMID: 36980286 PMCID: PMC10047773 DOI: 10.3390/cells12060944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Myelin basic protein (MBP) is one of the key structural elements of the myelin sheath and has autoantigenic properties in multiple sclerosis (MS). Its intracellular interaction network is still partially deconvoluted due to the unfolded structure, abnormally basic charge, and specific cellular localization. Here we used the fusion protein of MBP with TurboID, an engineered biotin ligase that uses ATP to convert biotin to reactive biotin-AMP that covalently attaches to nearby proteins, to determine MBP interactome. Despite evident benefits, the proximity labeling proteomics technique generates high background noise, especially in the case of proteins tending to semi-specific interactions. In order to recognize unique MBP partners, we additionally mapped protein interaction networks for deaminated MBP variant and cyclin-dependent kinase inhibitor 1 (p21), mimicking MBP in terms of natively unfolded state, size and basic amino acid clusters. We found that in the plasma membrane region, MBP is colocalized with adhesion proteins occludin and myelin protein zero-like protein 1, solute carrier family transporters ZIP6 and SNAT1, Eph receptors ligand Ephrin-B1, and structural components of the vesicle transport machinery-synaptosomal-associated protein 23 (SNAP23), vesicle-associated membrane protein 3 (VAMP3), protein transport protein hSec23B and cytoplasmic dynein 1 heavy chain 1. We also detected that MBP potentially interacts with proteins involved in Fe2+ and lipid metabolism, namely, ganglioside GM2 activator protein, long-chain-fatty-acid-CoA ligase 4 (ACSL4), NADH-cytochrome b5 reductase 1 (CYB5R1) and metalloreductase STEAP3. Assuming the emerging role of ferroptosis and vesicle cargo docking in the development of autoimmune neurodegeneration, MBP may recruit and regulate the activity of these processes, thus, having a more inclusive role in the integrity of the myelin sheath.
Collapse
Affiliation(s)
- Evgeniya V Smirnova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Tatiana V Rakitina
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Rustam H Ziganshin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - George A Saratov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (National Research University), 141701 Dolgoprudny, Russia
| | - Georgij P Arapidi
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (National Research University), 141701 Dolgoprudny, Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Alexey A Belogurov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Department of Biological Chemistry, Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of Russian Federation, 127473 Moscow, Russia
| | - Anna A Kudriaeva
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
7
|
Purazo ML, Ice RJ, Shimpi R, Hoenerhoff M, Pugacheva EN. NEDD9 Overexpression Causes Hyperproliferation of Luminal Cells and Cooperates with HER2 Oncogene in Tumor Initiation: A Novel Prognostic Marker in Breast Cancer. Cancers (Basel) 2023; 15:1119. [PMID: 36831460 PMCID: PMC9954084 DOI: 10.3390/cancers15041119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/23/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
HER2 overexpression occurs in 10-20% of breast cancer patients. HER2+ tumors are characterized by an increase in Ki67, early relapse, and increased metastasis. Little is known about the factors influencing early stages of HER2- tumorigenesis and diagnostic markers. Previously, it was shown that the deletion of NEDD9 in mouse models of HER2 cancer interferes with tumor growth, but the role of NEDD9 upregulation is currently unexplored. We report that NEDD9 is overexpressed in a significant subset of HER2+ breast cancers and correlates with a limited response to anti-HER2 therapy. To investigate the mechanisms through which NEDD9 influences HER2-dependent tumorigenesis, we generated MMTV-Cre-NEDD9 transgenic mice. The analysis of mammary glands shows extensive ductal epithelium hyperplasia, increased branching, and terminal end bud expansion. The addition of oncogene Erbb2 (neu) leads to the earlier development of early hyperplastic benign lesions (~16 weeks), with a significantly shorter latency than the control mice. Similarly, NEDD9 upregulation in MCF10A-derived acini leads to hyperplasia-like DCIS. This phenotype is associated with activation of ERK1/2 and AURKA kinases, leading to an increased proliferation of luminal cells. These findings indicate that NEDD9 is setting permissive conditions for HER2-induced tumorigenesis, thus identifying this protein as a potential diagnostic marker for early detection.
Collapse
Affiliation(s)
- Marc L. Purazo
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| | - Ryan J. Ice
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| | - Rahul Shimpi
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| | - Mark Hoenerhoff
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elena N. Pugacheva
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
- Department of Biochemistry & Molecular Medicine, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|
8
|
Feng J, Ouyang H, Wang J, Pan D, Sheng L, Xu C, Lin W, Hu D, Chang C, Jia D. MPZL1 upregulation promotes tumor metastasis and correlates with unfavorable prognosis in non-small cell lung cancer. Carcinogenesis 2022; 43:919-929. [PMID: 35727197 DOI: 10.1093/carcin/bgac055] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/18/2022] [Accepted: 06/16/2022] [Indexed: 01/13/2023] Open
Abstract
Non-small cell lung cancer (NSCLC), accounting for 85% of all lung cancer, is one of the leading causes of cancer-related death worldwide. Previously, we demonstrated that MPZL1 gene amplification promotes liver cancer metastasis through activating Src/Cortactin pathway. However, the clinical relevance and biological roles of the MPZL1 gene in lung cancer are still unknown. Here, we found that MPZL1 expression upregulates in human NSCLC, which is partly due to the copy number amplification of this gene. Next, we observed that high MPZL1 expression correlates with unfavorable prognosis of NSCLC patients. We further demonstrated that ectopic MPZL1 overexpression promotes in vitro migratory but not proliferation and colony formation abilities of both H1299 and H460 cells. Consistently, we found that MPZL1 knockdown impairs the migratory abilities of A549 and H1775 cells. Moreover, we found that MPZL1 knockdown inhibits in vivo metastatic but not tumor growth abilities of the A549 cells. Additionally, a total of 297 differentially expressed genes (DEGs) were identified by RNA sequencing in A549 cells upon MPZL1 knockdown. By integrative analysis of DEGs regulated by MPZL1 in A549 cells and human NSCLC tissues, we revealed that COL11A1 is the potential effector gene that positively regulated by MPZL1 and correlates with poor prognosis of NSCLC patients. In conclusion, our work indicates that one of the mechanisms by which MPZL1 promotes NSCLC metastasis is through upregulating the COL11A1, and MPZL1 can be used as a biomarker to predict the prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Jian Feng
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huiling Ouyang
- Laboratory of Cancer Genomics and Biology, Department of Urology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Deshen Pan
- Laboratory of Cancer Genomics and Biology, Department of Urology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luoyan Sheng
- Laboratory of Cancer Genomics and Biology, Department of Urology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaoliang Xu
- Laboratory of Cancer Genomics and Biology, Department of Urology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weihong Lin
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Dingzhong Hu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Chang
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Deshui Jia
- Laboratory of Cancer Genomics and Biology, Department of Urology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Ma J, Zhang L, Li S, Liu H. BRPCA: Bounded Robust Principal Component Analysis to Incorporate Similarity Network for N7-Methylguanosine(m 7G) Site-Disease Association Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:3295-3306. [PMID: 34469307 DOI: 10.1109/tcbb.2021.3109055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Recent studies have revealed that N7-methylguanosine(m7G) plays a pivotal role in various biological processes and disease pathogenesis. To date, transcriptome-wide m7G modification sites have been identified by high-throughput sequencing approaches, and some related information has been recorded in a few biological databases. However, the mechanism of site action in disease remains uncharted. Wet experiments can help identify true m7G sites with high confidence, but it is time-consuming to find the true ones in such a large number of sites, which will also cost too much. Thus, computational methods are emergently needed to predict the associations between m7G sites and various diseases, thus help to uncover potential active sites for specific diseases. In this article, we proposed a bounded robust principal component analysis (BRPCA) method to predict unknown m7G-disease association based on similarity information. Importantly, BRPCA tolerates the noise and redundancy existing in association and similarity information. Moreover, a suitable bounded constraint is incorporated into BRPCA to ensure that the predicted association scores locate in a meaningful interval. The extensive experiments demonstrate the superiority and robustness of the BRPCA.
Collapse
|
10
|
Novel Gene Signatures as Prognostic Biomarkers for Predicting the Recurrence of Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14040865. [PMID: 35205612 PMCID: PMC8870597 DOI: 10.3390/cancers14040865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary A high percentage of patients who undergo surgical resection for hepatocellular carcinoma (HCC) experience recurrence. Therefore, identification of accurate molecular markers for predicting recurrence of HCC is important. We analyzed recurrence and non-recurrence HCC tissues using two public omics datasets comprising microarray and RNA-sequencing and found novel gene signatures associated with recurrent HCC. These molecules might be used to not only predict for recurrence of HCC but also act as potential prognostic indicators for patients with HCC. Abstract Hepatocellular carcinoma (HCC) has a high rate of cancer recurrence (up to 70%) in patients who undergo surgical resection. We investigated prognostic gene signatures for predicting HCC recurrence using in silico gene expression analysis. Recurrence-associated gene candidates were chosen by a comparative analysis of gene expression profiles from two independent whole-transcriptome datasets in patients with HCC who underwent surgical resection. Five promising candidate genes, CETN2, HMGA1, MPZL1, RACGAP1, and SNRPB were identified, and the expression of these genes was evaluated using quantitative reverse transcription PCR in the validation set (n = 57). The genes CETN2, HMGA1, RACGAP1, and SNRPB, but not MPZL1, were upregulated in patients with recurrent HCC. In addition, the combination of HMGA1 and MPZL1 demonstrated the best area under the curve (0.807, 95% confidence interval [CI] = 0.681–0.899) for predicting HCC recurrence. In terms of clinicopathological correlation, CETN2, MPZL1, RACGAP1, and SNRPB were upregulated in patients with microvascular invasion, and the expression of MPZL1 and SNRPB was increased in proportion to the Edmonson tumor differentiation grade. Additionally, overexpression of CETN2, HMGA1, and RACGAP1 correlated with poor overall survival (OS) and disease-free survival (DFS) in the validation set. Finally, Cox regression analysis showed that the expression of serum alpha-fetoprotein and RACGAP1 significantly affected OS, whereas platelet count, microvascular invasion, and HMGA1 expression significantly affected DFS. In conclusion, HMGA1 and RACGAP1 may be potential prognostic biomarkers for predicting the recurrence of HCC after surgical resection.
Collapse
|
11
|
Kazemein Jasemi NS, Reza Ahmadian M. Allosteric regulation of GRB2 modulates RAS activation. Small GTPases 2022; 13:282-286. [PMID: 35703160 DOI: 10.1080/21541248.2022.2089001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
RAS activation is a multiple-step process in which linkage of the extracellular stimuli to the RAS activator SOS1 is the main step in RAS activation. GRB2 adaptor protein is the main modulator in SOS1 recruitment to the plasma membrane and its activation. This interaction is well studied but the exact mechanism of GRB2-SOS1 complex formation and SOS1 activation has yet remained obscure. Here, a new allosteric mechanism for the GRB2 regulation is described as a prerequisite for the modulation of SOS1 activation. This regulatory mechanism comprises a series of intramolecular interactions which are potentiated by GRB2 interaction with upstream ligands.Abbreviations: GRB2, growth factor receptor-bound protein 2; SOS1, son of sevenless 1; RAS, Rat Sarcoma; GEF, guanine nucleotide exchange factor; GAP, GTPase-activating protein; HER2, human epidermal growth factor receptor; SH3, SRC Homology 3; SH2, SRC Homology 2; PRD, proline-rich domain; PRM, proline-rich motif; PRP, proline-rich peptide; RTK, receptor tyrosine kinases.
Collapse
Affiliation(s)
- Neda S Kazemein Jasemi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitaetsstrasse 1, Building 22.03, 40255 Düsseldorf, Germany
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitaetsstrasse 1, Building 22.03, 40255 Düsseldorf, Germany
| |
Collapse
|
12
|
The intramolecular allostery of GRB2 governing its interaction with SOS1 is modulated by phosphotyrosine ligands. Biochem J 2021; 478:2793-2809. [PMID: 34232285 DOI: 10.1042/bcj20210105] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/01/2021] [Accepted: 07/07/2021] [Indexed: 02/04/2023]
Abstract
Growth factor receptor-bound protein 2 (GRB2) is a trivalent adaptor protein and a key element in signal transduction. It interacts via its flanking nSH3 and cSH3 domains with the proline-rich domain (PRD) of the RAS activator SOS1 and via its central SH2 domain with phosphorylated tyrosine residues of receptor tyrosine kinases (RTKs; e.g. HER2). The elucidation of structural organization and mechanistic insights into GRB2 interactions, however, remain challenging due to their inherent flexibility. This study represents an important advance in our mechanistic understanding of how GRB2 links RTKs to SOS1. Accordingly, it can be proposed that (1) HER2 pYP-bound SH2 potentiates GRB2 SH3 domain interactions with SOS1 (an allosteric mechanism); (2) the SH2 domain blocks cSH3, enabling nSH3 to bind SOS1 first before cSH3 follows (an avidity-based mechanism); and (3) the allosteric behavior of cSH3 to other domains appears to be unidirectional, although there is an allosteric effect between the SH2 and SH3 domains.
Collapse
|
13
|
Vemulapalli V, Chylek LA, Erickson A, Pfeiffer A, Gabriel KH, LaRochelle J, Subramanian K, Cao R, Stegmaier K, Mohseni M, LaMarche MJ, Acker MG, Sorger PK, Gygi SP, Blacklow SC. Time-resolved phosphoproteomics reveals scaffolding and catalysis-responsive patterns of SHP2-dependent signaling. eLife 2021; 10:64251. [PMID: 33755016 PMCID: PMC8024022 DOI: 10.7554/elife.64251] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/21/2021] [Indexed: 12/21/2022] Open
Abstract
SHP2 is a protein tyrosine phosphatase that normally potentiates intracellular signaling by growth factors, antigen receptors, and some cytokines, yet is frequently mutated in human cancer. Here, we examine the role of SHP2 in the responses of breast cancer cells to EGF by monitoring phosphoproteome dynamics when SHP2 is allosterically inhibited by SHP099. The dynamics of phosphotyrosine abundance at more than 400 tyrosine residues reveal six distinct response signatures following SHP099 treatment and washout. Remarkably, in addition to newly identified substrate sites on proteins such as occludin, ARHGAP35, and PLCγ2, another class of sites shows reduced phosphotyrosine abundance upon SHP2 inhibition. Sites of decreased phospho-abundance are enriched on proteins with two nearby phosphotyrosine residues, which can be directly protected from dephosphorylation by the paired SH2 domains of SHP2 itself. These findings highlight the distinct roles of the scaffolding and catalytic activities of SHP2 in effecting a transmembrane signaling response.
Collapse
Affiliation(s)
- Vidyasiri Vemulapalli
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, United States.,Department of Biological Chemistry & Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Lily A Chylek
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, United States
| | - Alison Erickson
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Anamarija Pfeiffer
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Khal-Hentz Gabriel
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, United States.,Department of Biological Chemistry & Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Jonathan LaRochelle
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, United States.,Department of Biological Chemistry & Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Kartik Subramanian
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, United States
| | - Ruili Cao
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, United States
| | - Kimberley Stegmaier
- Department of Pediatric Oncology, Dana Farber Cancer Institute, Boston, United States
| | - Morvarid Mohseni
- Novartis Institutes for Biomedical Research, Cambridge, United States
| | | | - Michael G Acker
- Novartis Institutes for Biomedical Research, Cambridge, United States
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Stephen C Blacklow
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, United States.,Department of Biological Chemistry & Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| |
Collapse
|
14
|
Zhang S, Zhang S, Wang H, Huang X, Wang J, Li J, Cheng D, Wang H, Lu D, Wang Y. Silencing myelin protein zero-like 1 expression suppresses cell proliferation and invasiveness of human glioma cells by inhibiting multiple cancer-associated signal pathways. JOURNAL OF NEURORESTORATOLOGY 2021. [DOI: 10.26599/jnr.2021.9040017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Glioma is the most common primary malignant tumor of the adult central nervous system. It has high morbidity and poor survival. Myelin protein zero-like protein 1 (MPZL1) is a cell surface glycoprotein that activates numerous adhesion-dependent signaling pathways. MPZL1 plays important roles in human cancers that include metastatic process; however, it is not clear if MPZL1 plays a role in human glioma. Therefore, this study aimed to determine if silencing MPZL1 impacted the cell proliferative features of human glioma cells. First, MPZL1 expression was investigated in human glioma samples and tumor cell lines. Then the effects of small interfering RNA (siRNA)-targeting MPZL1 were analyzed on proliferation, colony formation, cell cycle progression, and invasion of human glioma cells. The results from this study demonstrated that MPZL1 was highly expressed in human glioma tissues and glioma cell lines. In addition, knockdown of MPZL1 significantly inhibited cell proliferation, colony formation, and invasiveness of glioma cells, and effectively induced cell cycle arrest at the G1 phase. Western blotting analysis indicated that silencing MPZL1 expression downregulated the expression of matrix metalloproteinase-2 (MMP-2), WNT1, caspase-3, cyclin A1, epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), and upregulated p53. The results from this study suggest that MPZL1 might be a marker for tumors and could be a potential therapeutic target for human glioma.
Collapse
|
15
|
Tahir R, Renuse S, Udainiya S, Madugundu AK, Cutler JA, Nirujogi RS, Na CH, Xu Y, Wu X, Pandey A. Mutation-Specific and Common Phosphotyrosine Signatures of KRAS G12D and G13D Alleles. J Proteome Res 2020; 20:670-683. [PMID: 32986951 DOI: 10.1021/acs.jproteome.0c00587] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
KRAS is one of the most frequently mutated genes across all cancer subtypes. Two of the most frequent oncogenic KRAS mutations observed in patients result in glycine to aspartic acid substitution at either codon 12 (G12D) or 13 (G13D). Although the biochemical differences between these two predominant mutations are not fully understood, distinct clinical features of the resulting tumors suggest involvement of disparate signaling mechanisms. When we compared the global phosphotyrosine proteomic profiles of isogenic colorectal cancer cell lines bearing either G12D or G13D KRAS mutation, we observed both shared as well as unique signaling events induced by the two KRAS mutations. Remarkably, while the G12D mutation led to an increase in membrane proximal and adherens junction signaling, the G13D mutation led to activation of signaling molecules such as nonreceptor tyrosine kinases, MAPK kinases, and regulators of metabolic processes. The importance of one of the cell surface molecules, MPZL1, which was found to be hyperphosphorylated in G12D cells, was confirmed by cellular assays as its knockdown led to a decrease in proliferation of G12D but not G13D expressing cells. Overall, our study reveals important signaling differences across two common KRAS mutations and highlights the utility of our approach to systematically dissect subtle differences between related oncogenic mutants and potentially lead to individualized treatments.
Collapse
Affiliation(s)
- Raiha Tahir
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Santosh Renuse
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Savita Udainiya
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India.,Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India.,Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Anil K Madugundu
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Institute of Bioinformatics, International Technology Park, Bangalore 560066, India.,Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Jevon A Cutler
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Pre-Doctoral Training Program in Human Genetics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Raja Sekhar Nirujogi
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Chan Hyun Na
- Department of Neurology, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Yaoyu Xu
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Xinyan Wu
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Akhilesh Pandey
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India.,Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
16
|
Xu L, Zhou C, Pan R, Tang J, Wang J, Li B, Huang T, Duan S, Xu C. PTPN11 hypomethylation is associated with gastric cancer progression. Oncol Lett 2020; 19:1693-1700. [PMID: 32194661 PMCID: PMC7039138 DOI: 10.3892/ol.2020.11250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 10/14/2019] [Indexed: 11/06/2022] Open
Abstract
Protein tyrosine phosphatase non-receptor type 11 (PTPN11) encodes the tyrosine phosphatase SHP-2 that is overexpressed in gastric cancer (GC). In the present study, the association of PTPN11 methylation levels with the incidence of GC and its correlation with SHP-2 overexpression were investigated. The methylation levels of PTPN11 in tumor and adjacent normal tissues of 112 GC patients were assessed by quantitative methylation specific PCR (qMSP). The Cancer Genome Atlas (TCGA) public database was used to analyze the association between PTPN11 methylation and PTPN11 expression. Survival analyses were conducted in order to evaluate the prognostic value of PTPN11 methylation for GC. The results of the qMSP analysis indicated that the methylation levels of PTPN11 in GC tumor tissues were significantly decreased compared with those noted in the normal adjacent tissues (mean with standard deviation: 40.91±26.33 vs. 51.99±37.37, P=0.007). An inverse correlation between PTPN11 methylation levels and PTPN11 mRNA expression levels (P=4×10-6, r=-0.237) was noted. Subgroup analyses indicated that the association of PTPN11 hypomethylation with the incidence of GC was specific to male subjects (P=0.015), heavy drinking patients (P=0.019), patients with poor tumor differentiation (P=0.010) and patients with tumor node and metastasis (TNM) stage III+IV (P=0.008). Kaplan-Meier analyses and log-rank test suggested that PTPN11 hypomethylation was not associated with GC patient overall survival (P=0.605) and recurrence (P=0.485), although it could predict the recurrence of GC patients up to and including 60 years (≤60, P=0.049). The results indicated that PTPN11 levels were hypomethylated in GC patients. TCGA data analysis suggested that PTPN11 hypomethylation could cause an upregulation in the transcription levels of PTPN11. Although, this may explain the pattern of SHP-2 overexpression in GC, additional studies are required to verify this hypothesis. The association of PTPN11 hypomethylation with GC incidence may be specific to male patients, heavy drinking patients, patients with poor tumor differentiation and patients with TNM stage of III+IV. PTPN11 hypomethylation can be considered a biomarker for the recurrence of GC patients with an age of 60 years or lower.
Collapse
Affiliation(s)
- Lele Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215200, P.R. China
| | - Cong Zhou
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Ranran Pan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Junjian Tang
- Department of Vascular Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, P.R. China
| | - Jinzhi Wang
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Bin Li
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Tianyi Huang
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Shiwei Duan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215200, P.R. China
| |
Collapse
|
17
|
Chen D, Cao L, Wang X. MPZL1 promotes tumor cell proliferation and migration via activation of Src kinase in ovarian cancer. Oncol Rep 2019; 42:679-687. [PMID: 31233194 PMCID: PMC6610034 DOI: 10.3892/or.2019.7199] [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: 03/10/2018] [Accepted: 05/28/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor metastasis is the leading cause of mortality in patients with advanced ovarian cancer. Myelin protein zero like 1 (MPZL1) is a transmembrane glycoprotein that promotes migration of hepatocellular carcinoma cells and is involved in extracellular matrix-induced signal transduction. However, the functional role of MPZL1 in ovarian cancer has not been well elucidated. The present study conducted western blotting, phase-contrast imaging and immunohistochemistry to reveal the functions of MPZL1 in ovarian cancer. The present study demonstrated that the expression levels of MPZL1 were associated with malignant features of ovarian cancer. Furthermore, overexpression of MPZL1 significantly promoted cell proliferation, migration and invasion of ovarian cancer cells. Conversely, MPZL1 depletion by short hairpin RNA inhibited migration and invasion of ovarian cancer cells. In addition, this study demonstrated that phosphorylation of Src kinase was increased upon MPZL1 overexpression. Additionally, phosphorylation and activation of pro-metastatic proteins p130 and cortactin were induced by phosphorylated Src kinase. Collectively, these findings indicated that MPZL1 may be a novel pro-metastatic gene, which promotes tumor cell proliferation and migration through Src-mediated phosphorylation of p130 and cortactin in ovarian cancer.
Collapse
Affiliation(s)
- Danni Chen
- Department of Obstetrics and Gynecology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Lei Cao
- Department of Obstetrics and Gynecology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Xiaojie Wang
- Department of Obstetrics and Gynecology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| |
Collapse
|
18
|
Yu T, Liang L, Zhao X, Yin Y. Structural and biochemical studies of the extracellular domain of Myelin protein zero-like protein 1. Biochem Biophys Res Commun 2018; 506:883-890. [PMID: 30392906 DOI: 10.1016/j.bbrc.2018.10.161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/26/2018] [Indexed: 01/17/2023]
Abstract
Myelin protein zero-like protein 1 (MPZL1) is a member of the immunoglobulin superfamily, and is also a receptor of concanavalin A (ConA). MPZL1 is upregulated in hepatocellular carcinoma (HCC) and accelerates migration of HCC cells. However, function of MPZL1 as a receptor of ConA and its role in HCC development are largely unknown. To elucidate the functional basis, we have determined the crystal structure of the extracellular domain of MPZL1 at 2.7 Å resolution. Overall, it folds like a typical immunoglobulin variable-like domain that is much like MPZ. Unexpectedly, we found Asn50 is a unique glycosylation site and the glycosylation mediates its interaction with ConA. Furthermore, we also found that MPZL1 exists as a homodimer in the crystal, in which hydrogen bonds between Ser86 and Val145 play an important role. Our results demonstrate that glycosylation of Asn50 is essential for its function as a receptor of ConA. We propose that dimerization of MPZL1 participates in control of its signal transmission in cell adhesion.
Collapse
Affiliation(s)
- Tianshu Yu
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Ling Liang
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China; Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xuyang Zhao
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yuxin Yin
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China; Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China; Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
19
|
Jacquet K, Banerjee SL, Chartier FJM, Elowe S, Bisson N. Proteomic Analysis of NCK1/2 Adaptors Uncovers Paralog-specific Interactions That Reveal a New Role for NCK2 in Cell Abscission During Cytokinesis. Mol Cell Proteomics 2018; 17:1979-1990. [PMID: 30002203 DOI: 10.1074/mcp.ra118.000689] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 07/11/2018] [Indexed: 12/31/2022] Open
Abstract
Signals from cell surface receptors are often relayed via adaptor proteins. NCK1 and NCK2 are Src-Homology (SH) 2 and 3 domain adaptors that regulate processes requiring a remodeling of the actin cytoskeleton. Evidence from gene inactivation in mouse suggests that NCK1 and NCK2 are functionally redundant, although recent reports support the idea of unique functions for NCK1 and NCK2. We sought to examine this question further by delineating NCK1- and NCK2-specific signaling networks. We used both affinity purification-mass spectrometry and BioID proximity labeling to identify NCK1/2 signaling networks comprised of 98 proteins. Strikingly, we found 30 proteins restricted to NCK1 and 28 proteins specifically associated with NCK2, suggesting differences in their function. We report that Nck2 -/-, but not Nck1 -/- mouse embryo fibroblasts (MEFs) are multinucleated and display extended protrusions reminiscent of intercellular bridges, which correlate with an extended time spent in cytokinesis as well as a failure of a significant proportion of cells to complete abscission. Our data also show that the midbody of NCK2-deficient cells is not only increased in length, but also altered in composition, as judged by the mislocalization of AURKB, PLK1 and ECT2. Finally, we show that NCK2 function during cytokinesis requires its SH2 domain. Taken together, our data delineate the first high-confidence interactome for NCK1/2 adaptors and highlight several proteins specifically associated with either protein. Thus, contrary to what is generally accepted, we demonstrate that NCK1 and NCK2 are not completely redundant, and shed light on a previously uncharacterized function for the NCK2 adaptor protein in cell division.
Collapse
Affiliation(s)
- Kévin Jacquet
- From the ‡Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec G1R 2J6, QC, Canada.,§Centre de recherche sur le cancer de l'Université Laval, Québec G1R 2J6, QC, Canada.,¶PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec G1V 0A6, QC, Canada
| | - Sara L Banerjee
- From the ‡Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec G1R 2J6, QC, Canada.,§Centre de recherche sur le cancer de l'Université Laval, Québec G1R 2J6, QC, Canada.,¶PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec G1V 0A6, QC, Canada
| | - François J M Chartier
- From the ‡Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec G1R 2J6, QC, Canada.,§Centre de recherche sur le cancer de l'Université Laval, Québec G1R 2J6, QC, Canada.,¶PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec G1V 0A6, QC, Canada
| | - Sabine Elowe
- §Centre de recherche sur le cancer de l'Université Laval, Québec G1R 2J6, QC, Canada.,¶PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec G1V 0A6, QC, Canada.,‖Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Reproduction, santé de la mère et de l'enfant, Québec G1V 4G2, QC, Canada.,**Department of Pediatrics, Université Laval, Québec, QC, Canada
| | - Nicolas Bisson
- From the ‡Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec G1R 2J6, QC, Canada; .,§Centre de recherche sur le cancer de l'Université Laval, Québec G1R 2J6, QC, Canada.,¶PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec G1V 0A6, QC, Canada.,‡‡Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec G1V 0A6, QC, Canada
| |
Collapse
|