1
|
Farhadi P, Park T. The p130Cas-Crk/CrkL Axis: A Therapeutic Target for Invasive Cancers Unveiled by Collaboration Among p130Cas, Crk, and CrkL. Int J Mol Sci 2025; 26:4017. [PMID: 40362257 PMCID: PMC12071665 DOI: 10.3390/ijms26094017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Numerous studies have documented the involvement of p130Cas (Crk-associated substrate) in a wide range of cellular processes across different types of cells. These processes encompass cell transformation, the connection between the extracellular matrix and the actin cytoskeleton, cell migration and invasion, and cardiovascular development. Moreover, p130Cas has been associated with the regulation of various physiological processes, including mammary, bone, brain, muscle, and liver homeostasis. The diverse functions of p130Cas can be attributed to its possession of multiple protein-protein interaction domains, which sets it apart as a unique class of adaptor protein. It is well established that p130Cas interacts critically with the CT10 regulator of kinase (Crk) adaptor protein family members, including CrkII, CrkI, and Crk-like (CrkL), which is the basis for the naming of the Cas family. The Crk family proteins play a crucial role in integrating signals from various sources, such as growth factors, extracellular matrix molecules, bacterial pathogens, and apoptotic cells. An increasing body of evidence suggests that the dysregulation of Crk family proteins is linked to various human diseases, including cancer and increased susceptibility to pathogen infections. This review focuses primarily on the structural and functional aspects of the interaction between p130Cas and the Crk family proteins, providing insights into how these proteins regulate specific signaling events. Furthermore, we delve into the functions of p130Cas and the Crk family proteins in both normal and tumor cells to gain a comprehensive understanding of their collaborative roles in cellular physiology and pathology. This review demonstrates that tumor cell migration and invasion are the two cellular functions that have been studied the most for the p130Cas-Crk/CrkL axis. Understanding the tumor cell migration and invasion that require both p130Cas and Crk/CrkL is necessary to further evaluate the role of the p130Cas-Crk/CrkL axis in cancer. Establishing the contribution of the p130Cas-Crk/CrkL axis to cancer will facilitate the development of cancer drugs targeting the axis to inhibit cancer cell dissemination and improve patient outcomes.
Collapse
Affiliation(s)
- Pegah Farhadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 67155, Iran
| | - Taeju Park
- Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas City, MO 64108, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| |
Collapse
|
2
|
Kusano S, Ueno-Yokohata H, Hori M, Ishibashi T, Fujimura J, Shimizu T, Ohki K, Kiyokawa N. TCF3::ZNF384 induces steroid resistance in B-cell precursor acute lymphoblastic leukemia cells. Pediatr Int 2025; 67:e70078. [PMID: 40391410 DOI: 10.1111/ped.70078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 12/26/2024] [Accepted: 02/25/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND ZNF384 rearrangements (ZNF384-r) are associated with distinct subgroups of B-cell precursor acute lymphoblastic leukemia (BCP-ALL) and the mixed phenotype of acute leukemia. Types of BCP-ALL with ZNF384-r exhibit common immunophenotypic characteristics, whereas their clinical features are not uniform and TCF3::ZNF384-positive patients show a significantly poorer steroid response and higher frequency of relapse, while EP300::ZNF384-positive patients exhibit a favorable response to conventional chemotherapy. Therefore, we aimed to investigate the differences in biological effects between these two ZNF384-r molecules. METHOD We transduced BCP-ALL cell lines with both TCF3::ZNF384 and EP300::ZNF384 by retrovirus-mediated gene transduction, and examined the biological effects. RESULTS Flow cytometric analysis and RT-qPCR revealed down-regulation of CD10 in BCP-ALL cells after transduction with both TCF3::ZNF384 and EP300::ZNF384. The annexin-V binding apoptosis assay indicated that TCF3::ZNF384-, but not EP300::ZNF384-, expressing cells exhibited increased resistance to dexamethasone-induced apoptosis. By means of an oligonucleotide microarray and RT-qPCR, we observed that the transduction of TCF3::ZNF384, but not EP300::ZNF384, leads to significant enhancement of cyclin D2 (CCND2) gene expression in BCP-ALL cells, but no growth advantage was observed. CONCLUSION Our data suggest that the acquisition of dexamethasone resistance in BCP-ALL cell lines is an effect of TCF3::ZNF384 protein distinct from EP300::ZNF384. Other than the common functions of ZNF384-r that contribute to the development of leukemia with a lineage-ambiguous phenotype, TCF3::ZNF384 may exhibit a fusion partner-dependent function distinct from EP300::ZNF384 and participate in the formation of characteristic clinical features of TCF3::ZNF384-expressing ALL patients.
Collapse
Affiliation(s)
- Shinpei Kusano
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Setagaya-ku, Japan
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Hitomi Ueno-Yokohata
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Setagaya-ku, Japan
| | - Momoka Hori
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Setagaya-ku, Japan
- Graduate School of Engineering Science, Yokohama National University, Yokohama, Japan
| | - Takeshi Ishibashi
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Junya Fujimura
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Kentaro Ohki
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Setagaya-ku, Japan
| | - Nobutaka Kiyokawa
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Setagaya-ku, Japan
| |
Collapse
|
3
|
Yang Y, He R, Li D, Mu T, Kuang Z, Wang M. The pivotal role of ZNF384: driving the malignant behavior of serous ovarian cancer cells via the LIN28B/UBD axis. Cell Biol Toxicol 2024; 40:100. [PMID: 39562372 PMCID: PMC11576860 DOI: 10.1007/s10565-024-09938-6] [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: 02/28/2024] [Accepted: 10/16/2024] [Indexed: 11/21/2024]
Abstract
Zinc finger protein 384 (ZNF384) is a highly conserved transcribed gene associated with the development of multiple tumors, however, its role and mechanism in serous ovarian cancer (SOC) are unknown. We first confirmed that ZNF384 was abnormally highly expressed in SOC tissues by bioinformatics analysis and immunohistochemistry. We further used lentivirus packaging and transfection techniques to construct ZNF384 overexpression or knockdown cell lines, and through a series of cell function experiments, gradually verified that ZNF384 promoted a series of malignant behaviors of SOC cell proliferation, migration, and invasion. By establishing a xenotransplantation model in nude mice, it was confirmed that ZNF384 promoted the progress of SOC in vivo. Mechanistically, Overexpression of ZNF384 enhanced the transcriptional activity of Lin-28 homolog B (LIN28B), which promoted the malignant behavior of SOC cells. In addition, LIN28B could regulate the expression of the downstream factor ubiquitin D (UBD) in SOC cells, further promoting the development of SOC. This study shows that ZNF384 aggravates the malignant behavior of SOC cells through the LIN28B/UBD axis, which may be used as a diagnostic biomarker for patients with SOC.
Collapse
Affiliation(s)
- Ye Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Runze He
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Dongxiao Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Tianli Mu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Ziteng Kuang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Min Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
4
|
Yamada C, Okada K, Odaira K, Tokoro M, Iwamoto E, Sanada M, Noura M, Okamoto S, Yasuda T, Tsuzuki S, Kiyoi H, Hayakawa F. RGS1 and CREB5 are direct and common transcriptional targets of ZNF384-fusion proteins. Cancer Med 2024; 13:e7471. [PMID: 39015025 PMCID: PMC11252495 DOI: 10.1002/cam4.7471] [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: 01/18/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND ZNF384-fusion (Z-fusion) genes were recently identified in B-cell acute lymphoblastic leukemia (B-ALL) and are frequent in Japanese adult patients. The frequency is about 20% in those with Philadelphia chromosome-negative B-ALL. ZNF384 is a transcription factor and Z-fusion proteins have increased transcriptional activity; however, the detailed mechanisms of leukemogenesis of Z-fusion proteins have yet to be clarified. METHODS We established three transfectants of cell lines expressing different types of Z-fusion proteins, and analyzed their gene expression profile (GEP) by RNA-seq. We also analyzed the GEP of clinical ALL samples using our previous RNA-seq data of 323 Japanese ALL patients. We selected upregulated genes in both Z-fusion gene-expressing transfectants and Z-fusion gene-positive ALL samples, and investigated the binding of Z-fusion proteins to regulatory regions of the candidate genes by ChIP-qPCR. RESULTS We selected six commonly upregulated genes. After the investigation by ChIP-qPCR, we finally identified CREB5 and RGS1 as direct and common target genes. RGS1 is an inhibitor of CXCL12-CXCR4 signaling that is required for the homing of hematopoietic progenitor cells to the bone marrow microenvironment and development of B cells. Consistent with this, Z-fusion gene transfectants showed impaired migration toward CXCL12. CONCLUSIONS We identified CREB5 and RGS1 as direct and common transcriptional targets of Z-fusion proteins. The present results provide novel insight into the aberrant transcriptional regulation by Z-fusion proteins.
Collapse
Affiliation(s)
- Chiharu Yamada
- Division of Cellular and Genetic Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Kentaro Okada
- Division of Cellular and Genetic Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Koya Odaira
- Division of Cellular and Genetic Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Mahiru Tokoro
- Division of Cellular and Genetic Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Eisuke Iwamoto
- Clinical Research CenterNational Hospital Organization Nagoya Medical CenterNagoyaJapan
| | - Masashi Sanada
- Clinical Research CenterNational Hospital Organization Nagoya Medical CenterNagoyaJapan
| | - Mina Noura
- Division of Cellular and Genetic Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Syuichi Okamoto
- Division of Cellular and Genetic Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Takahiko Yasuda
- Clinical Research CenterNational Hospital Organization Nagoya Medical CenterNagoyaJapan
| | - Shinobu Tsuzuki
- Department of BiochemistryAichi Medical University School of MedicineNagakuteJapan
| | - Hitoshi Kiyoi
- Department of Hematology and OncologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Fumihiko Hayakawa
- Division of Cellular and Genetic Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
5
|
Wang Q, Han R, Xing H, Li H. A consensus genome of sika deer (Cervus nippon) and transcriptome analysis provided novel insights on the regulation mechanism of transcript factor in antler development. BMC Genomics 2024; 25:617. [PMID: 38890595 PMCID: PMC11186158 DOI: 10.1186/s12864-024-10522-9] [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: 01/15/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Sika deer (Cervus nippon) holds significance among cervids, with three genomes recently published. However, these genomes still contain hundreds of gaps and display significant discrepancies in continuity and accuracy. This poses challenges to functional genomics research and the selection of an appropriate reference genome. Thus, obtaining a high-quality reference genome is imperative to delve into functional genomics effectively. FINDINGS Here we report a high-quality consensus genome of male sika deer. All 34 chromosomes are assembled into single-contig pseudomolecules without any gaps, which is the most complete assembly. The genome size is 2.7G with 23,284 protein-coding genes. Comparative genomics analysis found that the genomes of sika deer and red deer are highly conserved, an approximately 2.4G collinear regions with up to 99% sequence similarity. Meanwhile, we observed the fusion of red deer's Chr23 and Chr4 during evolution, forming sika deer's Chr1. Additionally, we identified 607 transcription factors (TFs) that are involved in the regulation of antler development, including RUNX2, SOX6, SOX8, SOX9, PAX8, SIX2, SIX4, SIX6, SPI1, NFAC1, KLHL8, ZN710, JDP2, and TWST2, based on this consensus reference genome. CONCLUSIONS Our results indicated that we acquired a high-quality consensus reference genome. That provided valuable resources for understanding functional genomics. In addition, discovered the genetic basis of sika-red hybrid fertility and identified 607 significant TFs that impact antler development.
Collapse
Affiliation(s)
- Qianghui Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, China
| | - Ruobing Han
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, China
| | - Haihua Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, China
| | - Heping Li
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, China.
| |
Collapse
|
6
|
Kazemein Jasemi NS, Mehrabipour M, Magdalena Estirado E, Brunsveld L, Dvorsky R, Ahmadian MR. Functional Classification and Interaction Selectivity Landscape of the Human SH3 Domain Superfamily. Cells 2024; 13:195. [PMID: 38275820 PMCID: PMC10814857 DOI: 10.3390/cells13020195] [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: 12/12/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
SRC homology 3 (SH3) domains are critical interaction modules that orchestrate the assembly of protein complexes involved in diverse biological processes. They facilitate transient protein-protein interactions by selectively interacting with proline-rich motifs (PRMs). A database search revealed 298 SH3 domains in 221 human proteins. Multiple sequence alignment of human SH3 domains is useful for phylogenetic analysis and determination of their selectivity towards PRM-containing peptides (PRPs). However, a more precise functional classification of SH3 domains is achieved by constructing a phylogenetic tree only from PRM-binding residues and using existing SH3 domain-PRP structures and biochemical data to determine the specificity within each of the 10 families for particular PRPs. In addition, the C-terminal proline-rich domain of the RAS activator SOS1 covers 13 of the 14 recognized proline-rich consensus sequence motifs, encompassing differential PRP pattern selectivity among all SH3 families. To evaluate the binding capabilities and affinities, we conducted fluorescence dot blot and polarization experiments using 25 representative SH3 domains and various PRPs derived from SOS1. Our analysis has identified 45 interacting pairs, with binding affinities ranging from 0.2 to 125 micromolar, out of 300 tested and potential new SH3 domain-SOS1 interactions. Furthermore, it establishes a framework to bridge the gap between SH3 and PRP interactions and provides predictive insights into the potential interactions of SH3 domains with PRMs based on sequence specifications. This novel framework has the potential to enhance the understanding of protein networks mediated by SH3 domain-PRM interactions and be utilized as a general approach for other domain-peptide interactions.
Collapse
Affiliation(s)
- Neda S. Kazemein Jasemi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (N.S.K.J.); (M.M.); (R.D.)
| | - Mehrnaz Mehrabipour
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (N.S.K.J.); (M.M.); (R.D.)
| | - Eva Magdalena Estirado
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, P.O. Box 513, 5600MB Eindhoven, The Netherlands; (E.M.E.); (L.B.)
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, P.O. Box 513, 5600MB Eindhoven, The Netherlands; (E.M.E.); (L.B.)
| | - Radovan Dvorsky
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (N.S.K.J.); (M.M.); (R.D.)
| | - Mohammad R. Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (N.S.K.J.); (M.M.); (R.D.)
| |
Collapse
|
7
|
Cui J, Liu X, Dong W, Liu Y, Ruan X, Zhang M, Wang P, Liu L, Xue Y. SNORD17-mediated KAT6B mRNA 2'-O-methylation regulates vasculogenic mimicry in glioblastoma cells. Cell Biol Toxicol 2023; 39:2841-2860. [PMID: 37058271 DOI: 10.1007/s10565-023-09805-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/24/2023] [Indexed: 04/15/2023]
Abstract
Glioblastoma (GBM) is a primary tumor in the intracranial compartment. Vasculogenic mimicry (VM) is a process in which a pipeline of tumor cells that provide blood support to carcinogenic cells is formed, and studying VM could provide a new strategy for clinical targeted treatment of GBM. In the present study, we found that SNORD17 and ZNF384 were significantly upregulated and promoted VM in GBM, whereas KAT6B was downregulated and inhibited VM in GBM. RTL-P assays were performed to verify the 2'-O-methylation of KAT6B by SNORD17; IP assays were used to detect the acetylation of ZNF384 by KAT6B. In addition, the binding of ZNF384 to the promoter regions of VEGFR2 and VE-cadherin promoted transcription, as validated by chromatin immunoprecipitation and luciferase reporter assays. And finally, knockdown of SNORD17 and ZNF384 combined with KAT6B overexpression effectively reduced the xenograft tumor size, prolonged the survival time of nude mice and reduced the number of VM channels. This study reveals a novel mechanism of the SNORD17/KAT6B/ZNF384 axis in modulating VM development in GBM that may provide a new goal for the comprehensive treatment of GBM.
Collapse
Affiliation(s)
- Jingyi Cui
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Xiaobai Liu
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Weiwei Dong
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yunhui Liu
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Mengyang Zhang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Ping Wang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China.
| |
Collapse
|
8
|
Gonzalez-Avila G, Sommer B, Flores-Soto E, Aquino-Galvez A. Hypoxic Effects on Matrix Metalloproteinases' Expression in the Tumor Microenvironment and Therapeutic Perspectives. Int J Mol Sci 2023; 24:16887. [PMID: 38069210 PMCID: PMC10707261 DOI: 10.3390/ijms242316887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
The tumor microenvironment (TME) is characterized by an acidic pH and low oxygen concentrations. Hypoxia induces neoplastic cell evasion of the immune surveillance, rapid DNA repair, metabolic reprogramming, and metastasis, mainly as a response to the hypoxic inducible factors (HIFs). Likewise, cancer cells increase matrix metalloproteinases' (MMPs) expression in response to TME conditions, allowing them to migrate from the primary tumor to different tissues. Since HIFs and MMPs are augmented in the hypoxic TME, it is easy to consider that HIFs participate directly in their expression regulation. However, not all MMPs have a hypoxia response element (HRE)-HIF binding site. Moreover, different transcription factors and signaling pathways activated in hypoxia conditions through HIFs or in a HIF-independent manner participate in MMPs' transcription. The present review focuses on MMPs' expression in normal and hypoxic conditions, considering HIFs and a HIF-independent transcription control. In addition, since the hypoxic TME causes resistance to anticancer conventional therapy, treatment approaches using MMPs as a target alone, or in combination with other therapies, are also discussed.
Collapse
Affiliation(s)
- Georgina Gonzalez-Avila
- Laboratorio de Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico;
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico;
| | - Arnoldo Aquino-Galvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico;
| |
Collapse
|
9
|
Navarrete-López P, Maroto M, Pericuesta E, Fernández-González R, Lombó M, Ramos-Ibeas P, Gutiérrez-Adán A. Loss of the importin Kpna2 causes infertility in male mice by disrupting the translocation of testis-specific transcription factors. iScience 2023; 26:107134. [PMID: 37456838 PMCID: PMC10338237 DOI: 10.1016/j.isci.2023.107134] [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: 12/20/2022] [Revised: 03/16/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Karyopherins mediate the movement between the nucleus and cytoplasm of specific proteins in diverse cellular processes. Through a loss-of-function approach, we here examine the role of Karyopherin Subunit Alpha 2 (Kpna2) in spermatogenesis. Knockout male mice exhibited reduced body size and sperm motility, increased sperm abnormalities, and led to the dysregulation of testis gene expression and ultimately to infertility. Impaired mRNA expression mainly affected clusters of genes expressed in spermatids and spermatocytes. Downregulated genes included a set of genes that participate in cell adhesion and extracellular matrix (ECM) organization. We detected both the enrichment of some transcription factors that bind to regions around transcription start sites of downregulated genes and the impaired transport of specific factors to the nucleus of spermatid cells. We propose that Kpna2 is essential in the seminiferous tubules for promoting the translocation of testis-specific transcription factors that control the expression of genes related to ECM organization.
Collapse
Affiliation(s)
| | - María Maroto
- Department of Animal Reproduction, INIA-CSIC, 28040 Madrid, Spain
| | - Eva Pericuesta
- Department of Animal Reproduction, INIA-CSIC, 28040 Madrid, Spain
| | | | - Marta Lombó
- Department of Animal Reproduction, INIA-CSIC, 28040 Madrid, Spain
| | | | | |
Collapse
|
10
|
Korff C, Atkinson E, Adaway M, Klunk A, Wek RC, Vashishth D, Wallace JM, Anderson-Baucum EK, Evans-Molina C, Robling AG, Bidwell JP. NMP4, an Arbiter of Bone Cell Secretory Capacity and Regulator of Skeletal Response to PTH Therapy. Calcif Tissue Int 2023; 113:110-125. [PMID: 37147466 PMCID: PMC10330242 DOI: 10.1007/s00223-023-01088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/21/2023] [Indexed: 05/07/2023]
Abstract
The skeleton is a secretory organ, and the goal of some osteoporosis therapies is to maximize bone matrix output. Nmp4 encodes a novel transcription factor that regulates bone cell secretion as part of its functional repertoire. Loss of Nmp4 enhances bone response to osteoanabolic therapy, in part, by increasing the production and delivery of bone matrix. Nmp4 shares traits with scaling factors, which are transcription factors that influence the expression of hundreds of genes to govern proteome allocation for establishing secretory cell infrastructure and capacity. Nmp4 is expressed in all tissues and while global loss of this gene leads to no overt baseline phenotype, deletion of Nmp4 has broad tissue effects in mice challenged with certain stressors. In addition to an enhanced response to osteoporosis therapies, Nmp4-deficient mice are less sensitive to high fat diet-induced weight gain and insulin resistance, exhibit a reduced disease severity in response to influenza A virus (IAV) infection, and resist the development of some forms of rheumatoid arthritis. In this review, we present the current understanding of the mechanisms underlying Nmp4 regulation of the skeletal response to osteoanabolics, and we discuss how this unique gene contributes to the diverse phenotypes among different tissues and stresses. An emerging theme is that Nmp4 is important for the infrastructure and capacity of secretory cells that are critical for health and disease.
Collapse
Affiliation(s)
- Crystal Korff
- Department of Medical and Molecular Genetics, Indiana University School of Medicine (IUSM), Indianapolis, IN, 46202, USA
| | - Emily Atkinson
- Department of Anatomy, Cell Biology & Physiology, IUSM, Indianapolis, IN, 46202, USA
| | - Michele Adaway
- Department of Anatomy, Cell Biology & Physiology, IUSM, Indianapolis, IN, 46202, USA
| | - Angela Klunk
- Department of Anatomy, Cell Biology & Physiology, IUSM, Indianapolis, IN, 46202, USA
| | - Ronald C Wek
- Department of Biochemistry and Molecular Biology, IUSM, Indianapolis, IN, USA
| | - Deepak Vashishth
- Center for Biotechnology & Interdisciplinary Studies and Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Joseph M Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, IUSM, Indianapolis, IN, USA
| | - Emily K Anderson-Baucum
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, IUSM, Indianapolis, IN, USA
| | - Carmella Evans-Molina
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, IUSM, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Disease and the Wells Center for Pediatric Research, IUSM, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA
- Department of Medicine, IUSM, Indianapolis, IN, USA
| | - Alexander G Robling
- Department of Anatomy, Cell Biology & Physiology, IUSM, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, IUSM, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA
| | - Joseph P Bidwell
- Department of Anatomy, Cell Biology & Physiology, IUSM, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, IUSM, Indianapolis, IN, USA.
| |
Collapse
|
11
|
D’Agostino C, Parisis D, Chivasso C, Hajiabbas M, Soyfoo MS, Delporte C. Aquaporin-5 Dynamic Regulation. Int J Mol Sci 2023; 24:ijms24031889. [PMID: 36768212 PMCID: PMC9915196 DOI: 10.3390/ijms24031889] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/21/2023] Open
Abstract
Aquaporin-5 (AQP5), belonging to the aquaporins (AQPs) family of transmembrane water channels, facilitates osmotically driven water flux across biological membranes and the movement of hydrogen peroxide and CO2. Various mechanisms have been shown to dynamically regulate AQP5 expression, trafficking, and function. Besides fulfilling its primary water permeability function, AQP5 has been shown to regulate downstream effectors playing roles in various cellular processes. This review provides a comprehensive overview of the current knowledge of the upstream and downstream effectors of AQP5 to gain an in-depth understanding of the physiological and pathophysiological processes involving AQP5.
Collapse
Affiliation(s)
- Claudia D’Agostino
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Dorian Parisis
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium
- Rheumatology Department, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium
| | - Clara Chivasso
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Maryam Hajiabbas
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Muhammad Shahnawaz Soyfoo
- Rheumatology Department, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium
| | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium
- Correspondence:
| |
Collapse
|
12
|
Charakopoulos E, Diamantopoulos PT, Zervakis K, Giannakopoulou N, Psichogiou M, Viniou NA. A case report of a fulminant Aeromonas hydrophila soft tissue infection in a patient with acute lymphoblastic leukemia harboring a rare translocation. Curr Med Res Opin 2022; 38:1125-1132. [PMID: 35575163 DOI: 10.1080/03007995.2022.2078079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
INTRODUCTION Aeromonads are gram-negative opportunistic bacteria, mainly found in aquatic environments. Hematologic patients are particularly at risk of Aeromonas soft tissue infections and septicemia, especially during chemotherapy-induced neutropenia. CASE DESCRIPTION A 46-year-old man was diagnosed with acute lymphoblastic leukemia characterized by the rare t(12;17)(p13;q21)/TAF15-ZNF384 aberration. On day 22 of chemotherapy, he developed febrile neutropenia followed by necrotizing fasciitis in his upper right extremity. Despite appropriate antibiotic therapy and prompt surgical intervention, he died within 36 h after the appearance of a fever. A multi-sensitive Aeromonas hydrophila was isolated from all cultural sites. DISCUSSION AND CONCLUSIONS In a previous paper we characterized the patient's aberration with cytogenetic and FISH analysis. Here, we provide details regarding the patient's rapidly progressing infection and underline the importance of maintaining high clinical suspicion of Aeromonas infections in acute leukemia. Given the unusually rapid progression of an infection caused by a rare non-resistant pathogen, and after considering data on the implication of metalloproteinase function in immune system regulation, a correlation between risk of severe infection and TAF15-ZNF384 aberrated acute lymphoblastic leukemia cannot be ruled out.
Collapse
Affiliation(s)
- Emmanouil Charakopoulos
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Greece
| | - Panagiotis T Diamantopoulos
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Greece
| | - Konstantinos Zervakis
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Greece
| | - Nefeli Giannakopoulou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Greece
| | - Mina Psichogiou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Greece
| | - Nora-Athina Viniou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Greece
| |
Collapse
|
13
|
de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, Devel L, Dufour A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol Rev 2022; 74:712-768. [PMID: 35738680 DOI: 10.1124/pharmrev.121.000349] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The first matrix metalloproteinase (MMP) was discovered in 1962 from the tail of a tadpole by its ability to degrade collagen. As their name suggests, matrix metalloproteinases are proteases capable of remodeling the extracellular matrix. More recently, MMPs have been demonstrated to play numerous additional biologic roles in cell signaling, immune regulation, and transcriptional control, all of which are unrelated to the degradation of the extracellular matrix. In this review, we will present milestones and major discoveries of MMP research, including various clinical trials for the use of MMP inhibitors. We will discuss the reasons behind the failures of most MMP inhibitors for the treatment of cancer and inflammatory diseases. There are still misconceptions about the pathophysiological roles of MMPs and the best strategies to inhibit their detrimental functions. This review aims to discuss MMPs in preclinical models and human pathologies. We will discuss new biochemical tools to track their proteolytic activity in vivo and ex vivo, in addition to future pharmacological alternatives to inhibit their detrimental functions in diseases. SIGNIFICANCE STATEMENT: Matrix metalloproteinases (MMPs) have been implicated in most inflammatory, autoimmune, cancers, and pathogen-mediated diseases. Initially overlooked, MMP contributions can be both beneficial and detrimental in disease progression and resolution. Thousands of MMP substrates have been suggested, and a few hundred have been validated. After more than 60 years of MMP research, there remain intriguing enigmas to solve regarding their biological functions in diseases.
Collapse
Affiliation(s)
- Luiz G N de Almeida
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Hayley Thode
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Yekta Eslambolchi
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sameeksha Chopra
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Daniel Young
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sean Gill
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Laurent Devel
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Antoine Dufour
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| |
Collapse
|
14
|
Dickerson KM, Qu C, Gao Q, Iacobucci I, Gu Z, Yoshihara H, Backhaus EA, Chang Y, Janke LJ, Xu B, Wu G, Papachristou EK, D'Santos CS, Roberts KG, Mullighan CG. ZNF384 fusion oncoproteins drive lineage aberrancy in acute leukemia. Blood Cancer Discov 2022; 3:240-263. [PMID: 35247902 DOI: 10.1158/2643-3230.bcd-21-0163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/21/2021] [Accepted: 02/28/2022] [Indexed: 11/16/2022] Open
Abstract
ZNF384-rearranged fusion oncoproteins (FO) define a subset of lineage ambiguous leukemias, but their mechanistic role in leukemogenesis and lineage ambiguity is poorly understood. Using viral expression in mouse and human hematopoietic stem and progenitor cells (HSPCs) and a Ep300::Znf384 knockin mouse model, we show that ZNF384 FO promote hematopoietic expansion, myeloid lineage skewing, and self-renewal. In mouse HSPCs, concomitant lesions, such as NRASG12D, were required for fully penetrant leukemia, whereas in human HSPCs expression of ZNF384 FO drove B/myeloid leukemia, with sensitivity of a ZNF384-rearranged xenograft to FLT3 inhibition in vivo. Mechanistically, ZNF384 FO occupy a subset of predominantly intragenic/enhancer regions with increased histone 3 lysine acetylation and deregulate expression of hematopoietic stem cell transcription factors. These data define a paradigm for FO-driven lineage ambiguous leukemia, in which expression in HSPCs results in deregulation of lineage-specific genes and hematopoietic skewing, progressing to full leukemia in the context of proliferative stress.
Collapse
Affiliation(s)
| | - Chunxu Qu
- St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Qingsong Gao
- St. Jude Children's Research Hospital, Memphis, United States
| | - Ilaria Iacobucci
- St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Zhaohui Gu
- City Of Hope National Medical Center, United States
| | | | - Emily A Backhaus
- St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Yunchao Chang
- St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Laura J Janke
- St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Beisi Xu
- St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Gang Wu
- St. Jude Children's Research Hospital, Memphis, United States
| | | | - Clive S D'Santos
- Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
| | | | | |
Collapse
|
15
|
Lejman M, Chałupnik A, Chilimoniuk Z, Dobosz M. Genetic Biomarkers and Their Clinical Implications in B-Cell Acute Lymphoblastic Leukemia in Children. Int J Mol Sci 2022; 23:2755. [PMID: 35269896 PMCID: PMC8911213 DOI: 10.3390/ijms23052755] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 02/04/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a heterogeneous group of hematologic malignancies characterized by abnormal proliferation of immature lymphoid cells. It is the most commonly diagnosed childhood cancer with an almost 80% cure rate. Despite favorable survival rates in the pediatric population, a significant number of patients develop resistance to therapy, resulting in poor prognosis. ALL is a heterogeneous disease at the genetic level, but the intensive development of sequencing in the last decade has made it possible to broaden the study of genomic changes. New technologies allow us to detect molecular changes such as point mutations or to characterize epigenetic or proteomic profiles. This process made it possible to identify new subtypes of this disease characterized by constellations of genetic alterations, including chromosome changes, sequence mutations, and DNA copy number alterations. These genetic abnormalities are used as diagnostic, prognostic and predictive biomarkers that play an important role in earlier disease detection, more accurate risk stratification, and treatment. Identification of new ALL biomarkers, and thus a greater understanding of their molecular basis, will lead to better monitoring of the course of the disease. In this article, we provide an overview of the latest information on genomic alterations found in childhood ALL and discuss their impact on patients' clinical outcomes.
Collapse
Affiliation(s)
- Monika Lejman
- Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Aleksandra Chałupnik
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (A.C.); (Z.C.); (M.D.)
| | - Zuzanna Chilimoniuk
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (A.C.); (Z.C.); (M.D.)
| | - Maciej Dobosz
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (A.C.); (Z.C.); (M.D.)
| |
Collapse
|
16
|
ZNF384 rearrangement in acute lymphocytic leukemia with renal involvement as the first manifestation is associated with a poor prognosis: a case report. Mol Cytogenet 2022; 15:4. [PMID: 35164825 PMCID: PMC8842518 DOI: 10.1186/s13039-022-00583-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/28/2022] [Indexed: 11/27/2022] Open
Abstract
Background Novel fusion genes such as ZNF384, have been identified in B-cell precursor acute lymphoblastic leukemia (BCP-ALL) in recent years. Patients harboring ZNF384 rearrangements have a distinctive immunophenotype with weak CD10 and aberrant CD13 and/or CD33 expression. Thus, ZNF384-rearranged ALL is a unique subtype of BCP-ALL. However, research on the prognostic significance of ZNF384 rearrangements has been limited to date, especially in adolescents. Case presentation We described a 17-year-old adolescent who was diagnosed with ALL and had renal involvement as the first manifestation, which was very rare in the existing studies. FISH analysis indicated a rearrangement of ZNF384 according to its probe. The patient had a typical characteristic immunophenotype of ZNF384 rearrangement, with CD10 negativity and CD13 and CD33 positivity. She had an unfavorable prognosis because she responded poorly to chemotherapy and developed a relapse shortly after reaching CR. Conclusion The importance of ZNF384 rearrangements in terms of prognosis remains unclear. We reported an adolescent who was diagnosed with ZNF384-rearranged ALL with renal involvement. She underwent different therapies, but her prognosis remained poor. Since ZNF384 rearrangements may act as a prognostic predictor in children or adolescents, early detection based on its characteristic immunophenotype is of great necessity. Supplementary Information The online version contains supplementary material available at 10.1186/s13039-022-00583-4.
Collapse
|
17
|
Yan Z, Zhou Y, Yang Y, Zeng C, Li P, Tian H, Tang X, Zhang G. Zinc finger protein 384 enhances colorectal cancer metastasis by upregulating MMP2. Oncol Rep 2022; 47:49. [PMID: 35029289 PMCID: PMC8771166 DOI: 10.3892/or.2022.8260] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Zinc finger proteins (ZNFs) serve key roles in tumor formation and progression; however, the functions and underlying mechanisms of dysregulated ZNF384 in colorectal cancer (CRC) are yet to be fully elucidated. Therefore, the present study initially aimed to investigate the expression levels of ZNF384 in CRC samples. Moreover, lentiviral ZNF384 overexpression and ZNF384 knockdown models were established in CRC cells. Transwell, wound healing and in vivo tail vein metastasis assays were carried out to evaluate the effects of ZNF384 on CRC cell metastasis. Furthermore, reverse transcription-quantitative PCR, western blotting, serial deletion, site-directed mutagenesis, dual-luciferase reporter and chromatin immunoprecipitation assays were conducted to investigate the potential underlying mechanisms. The results of the present study demonstrated that ZNF384 expression was markedly increased in CRC samples and this was associated with a poor prognosis. Notably, ZNF384 overexpression increased the levels of CRC cell invasion and migration, whereas ZNF384 knockdown inhibited CRC development. Moreover, the results of the present study demonstrated that ZNF384 mediated the expression of MMP2. MMP2 knockdown inhibited ZNF384-mediated CRC cell invasion and migration, whereas MMP2 overexpression ameliorated ZNF384 knockdown-induced inhibition of CRC progression. In addition, the results of the present study demonstrated that hypoxia-inducible factor 1α (HIF-1α) had the ability to bind to the ZNF384 promoter, thereby initiating ZNF384 expression. In human-derived CRC samples, the expression levels of ZNF384 were positively correlated with both MMP2 and HIF-1α expression. Collectively, these findings highlighted that ZNF384 may act as a prognostic marker and regulator of CRC metastasis.
Collapse
Affiliation(s)
- Zaihua Yan
- Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yu Zhou
- Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yang Yang
- Department of Dermatology, Xining First People's Hospital, Xining, Qinghai 810000, P.R. China
| | - Chongpu Zeng
- Department of General Surgery, Wangcang County People's Hospital, Guangyuan, Sichuan 628200, P.R. China
| | - Peidong Li
- Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Hongpeng Tian
- Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xuegui Tang
- Anorectal Department of Integrated Traditional Chinese and Western Medicine, The Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Guangjun Zhang
- Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
18
|
Singh JK, Smith R, Rother MB, de Groot AJL, Wiegant WW, Vreeken K, D’Augustin O, Kim RQ, Qian H, Krawczyk PM, González-Prieto R, Vertegaal ACO, Lamers M, Huet S, van Attikum H. Zinc finger protein ZNF384 is an adaptor of Ku to DNA during classical non-homologous end-joining. Nat Commun 2021; 12:6560. [PMID: 34772923 PMCID: PMC8589989 DOI: 10.1038/s41467-021-26691-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/19/2021] [Indexed: 12/14/2022] Open
Abstract
DNA double-strand breaks (DSBs) are among the most deleterious types of DNA damage as they can lead to mutations and chromosomal rearrangements, which underlie cancer development. Classical non-homologous end-joining (cNHEJ) is the dominant pathway for DSB repair in human cells, involving the DNA-binding proteins XRCC6 (Ku70) and XRCC5 (Ku80). Other DNA-binding proteins such as Zinc Finger (ZnF) domain-containing proteins have also been implicated in DNA repair, but their role in cNHEJ remained elusive. Here we show that ZNF384, a member of the C2H2 family of ZnF proteins, binds DNA ends in vitro and is recruited to DSBs in vivo. ZNF384 recruitment requires the poly(ADP-ribosyl) polymerase 1 (PARP1)-dependent expansion of damaged chromatin, followed by binding of its C2H2 motifs to the exposed DNA. Moreover, ZNF384 interacts with Ku70/Ku80 via its N-terminus, thereby promoting Ku70/Ku80 assembly and the accrual of downstream cNHEJ factors, including APLF and XRCC4/LIG4, for efficient repair at DSBs. Altogether, our data suggest that ZNF384 acts as a 'Ku-adaptor' that binds damaged DNA and Ku70/Ku80 to facilitate the build-up of a cNHEJ repairosome, highlighting a role for ZNF384 in DSB repair and genome maintenance.
Collapse
Affiliation(s)
- Jenny Kaur Singh
- grid.10419.3d0000000089452978Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Rebecca Smith
- grid.410368.80000 0001 2191 9284Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes)—UMR 6290, BIOSIT–UMS3480, F-35000 Rennes, France
| | - Magdalena B. Rother
- grid.10419.3d0000000089452978Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Anton J. L. de Groot
- grid.10419.3d0000000089452978Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Wouter W. Wiegant
- grid.10419.3d0000000089452978Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Kees Vreeken
- grid.10419.3d0000000089452978Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ostiane D’Augustin
- grid.410368.80000 0001 2191 9284Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes)—UMR 6290, BIOSIT–UMS3480, F-35000 Rennes, France ,grid.457349.80000 0004 0623 0579Institut de Biologie François Jacob, Institute of Cellular and Molecular Radiobiology, Université Paris-Saclay, Université de Paris, CEA, F-92265 Fontenay-aux-Roses, France
| | - Robbert Q. Kim
- grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Haibin Qian
- grid.16872.3a0000 0004 0435 165XDepartment of Medical Biology, Amsterdam University Medical Centers (location AMC), Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Przemek M. Krawczyk
- grid.16872.3a0000 0004 0435 165XDepartment of Medical Biology, Amsterdam University Medical Centers (location AMC), Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Román González-Prieto
- grid.16872.3a0000 0004 0435 165XDepartment of Medical Biology, Amsterdam University Medical Centers (location AMC), Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Alfred C. O. Vertegaal
- grid.16872.3a0000 0004 0435 165XDepartment of Medical Biology, Amsterdam University Medical Centers (location AMC), Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Meindert Lamers
- grid.16872.3a0000 0004 0435 165XDepartment of Medical Biology, Amsterdam University Medical Centers (location AMC), Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sébastien Huet
- grid.410368.80000 0001 2191 9284Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes)—UMR 6290, BIOSIT–UMS3480, F-35000 Rennes, France ,grid.440891.00000 0001 1931 4817Institut Universitaire de France, F-75000 Paris, France
| | - Haico van Attikum
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
19
|
Centonze G, Natalini D, Salemme V, Costamagna A, Cabodi S, Defilippi P. p130Cas/ BCAR1 and p140Cap/ SRCIN1 Adaptors: The Yin Yang in Breast Cancer? Front Cell Dev Biol 2021; 9:729093. [PMID: 34708040 PMCID: PMC8542790 DOI: 10.3389/fcell.2021.729093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
p130Cas/BCAR1 is an adaptor protein devoid of any enzymatic or transcriptional activity, whose modular structure with various binding motifs, allows the formation of multi-protein signaling complexes. This results in the induction and/or maintenance of signaling pathways with pleiotropic effects on cell motility, cell adhesion, cytoskeleton remodeling, invasion, survival, and proliferation. Deregulation of p130Cas/BCAR1 adaptor protein has been extensively demonstrated in a variety of human cancers in which overexpression of p130Cas/BCAR1 correlates with increased malignancy. p140Cap (p130Cas associated protein), encoded by the SRCIN1 gene, has been discovered by affinity chromatography and mass spectrometry analysis of putative interactors of p130Cas. It came out that p140Cap associates with p130Cas not directly but through its interaction with the Src Kinase. p140Cap is highly expressed in neurons and to a lesser extent in epithelial tissues such as the mammary gland. Strikingly, in vivo and in vitro analysis identified its tumor suppressive role in breast cancer and in neuroblastoma, showing an inverse correlation between p140Cap expression in tumors and tumor progression. In this review, a synopsis of 15 years of research on the role of p130Cas/BCAR1 and p140Cap/SRCIN1 in breast cancer will be presented.
Collapse
Affiliation(s)
- Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Andrea Costamagna
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Sara Cabodi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| |
Collapse
|
20
|
Jimi E, Honda H, Nakamura I. The unique function of p130Cas in regulating the bone metabolism. Pharmacol Ther 2021; 230:107965. [PMID: 34391790 DOI: 10.1016/j.pharmthera.2021.107965] [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: 11/30/2020] [Accepted: 07/20/2021] [Indexed: 11/19/2022]
Abstract
p130 Crk-associated substrate (Cas) functions as an adapter protein and plays important roles in certain cell functions, such as cell proliferation, spreading, migration, and invasion. Furthermore, it has recently been reported to have a new function as a mechanosensor. Since bone is a tissue that is constantly under gravity, it is exposed to mechanical stress. Mechanical unloading, such as in a microgravity environment in space or during bed rest, leads to a decrease in bone mass because of the suppression of bone formation and the stimulation of bone resorption. Osteoclasts are multinucleated bone-resorbing giant cells that recognize bone and then form a ruffled border in the resorption lacuna. p130Cas is a molecule located downstream of c-Src that is important for the formation of a ruffled border in osteoclasts. Indeed, osteoclast-specific p130Cas-deficient mice exhibit osteopetrosis due to osteoclast dysfunction, similar to c-Src-deficient mice. Osteoblasts subjected to mechanical stress induce both the phosphorylation of p130Cas and osteoblast differentiation. In osteocytes, mechanical stress regulates bone mass by shuttling p130Cas between the cytoplasm and nucleus. Oral squamous cell carcinoma (OSCC) cells express p130Cas more strongly than epithelial cells in normal tissues. The knockdown of p130Cas in OSCC cells suppressed the cell growth, the expression of receptor activator of NF-κB ligand, which induces osteoclast formation, and bone invasion by OSCC. Taken together, these findings suggest that p130Cas might be a novel therapeutic target molecule in bone diseases, such as osteoporosis, rheumatoid arthritis, bone loss due to bed rest, and bone invasion and metastasis of cancer.
Collapse
Affiliation(s)
- Eijiro Jimi
- Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Ichiro Nakamura
- Department of Rehabilitation, Yugawara Hospital, Japan Community Health Care Organization, 2-21-6 Chuo, Yugawara, Ashigara-shimo, Kanagawa 259-0396, Japan
| |
Collapse
|
21
|
Lin N, Yan X, Cai D, Wang L. Leukemia With TCF3-ZNF384 Rearrangement as a Distinct Subtype of Disease With Distinct Treatments: Perspectives From A Case Report and Literature Review. Front Oncol 2021; 11:709036. [PMID: 34395283 PMCID: PMC8357369 DOI: 10.3389/fonc.2021.709036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background ZNF384 rearrangements are found in 5-10% of B-cell acute lymphoblastic leukemia (B-ALL) and 48% of B cell/myeloid mixed phenotype acute leukemia (B/M MPAL). ZNF384-rearranged B-ALL is prone to lineage conversion after chemotherapy. TCF3 is the second most common rearrangement partner of ZNF384 in B-ALL (27.5%) and the most common partner in B/M MPAL (53.3%). TCF3-ZNF384 fusion is related to a poor steroid response and a high frequency of relapse. It is mostly reported in children and adolescents but rarely seen in adults. Patients and Methods Here, we report a rare case of adult common B-ALL with TCF3-ZNF384 fusion in which the patient relapsed after one cycle of consolidation chemotherapy. Relapsed leukemia cells from the bone marrow were cultured for 72 hours ex vivo, and a panel of 156 kinds of cytotoxic drugs, targeted therapy drugs, combination chemotherapy drugs, etc., was used for sensitivity screening. The literature on TCF3-ZNF384 fusion was reviewed, and reported cases with TCF3-ZNF384 fusion were summarized. Clinical characteristics were compared between B-ALL and MPAL with TCF3-ZNF384 fusion. Results The relapsed lymphoblasts showed moderate sensitivity to both acute myelocytic leukemia (AML) - and acute lymphoblastic leukemia (ALL)-directed combination chemotherapy schemes, as well as to multiple targeted therapeutic drugs. The hyper-CVAD (B) scheme showed synergistic effects with multiple targeted compounds and had the highest sensitivity. The patient chose the hyper-CVAD (B) scheme combined with sorafenib and achieved complete remission (CR), then consolidated with myeloid-directed homoharringtonine+cytarabine+daunorubicin (HAD) scheme and gained molecular CR. By reviewing the literature, we found that both the genomic landscapes and gene expression profiles of ZNF384-rearranged B-ALL and MPAL are similar and that both diseases have lineage plasticity. The gene expression profile in TCF3-ZNF384-positive patients shows enrichment of hematopoietic stem cell features. No significant differences in clinical characteristics were found between TCF3-ZNF384-positive ALL and MPAL. Conclusion TCF3-ZNF384-positive leukemia may be a distinct subtype of leukemia regardless of immunophenotype. Considering the frequent lineage switches and sensitivity to both ALL- and AML-directed schemes, a uniform strategy directed at both lymphoid and myeloid lineages or at hematopoietic stem cells may be better for TCF3-ZNF384-positive leukemia. Small molecule targeted therapies may be promising treatment options and deserve further investigation.
Collapse
Affiliation(s)
- Na Lin
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaojing Yan
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Dali Cai
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Lei Wang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Yaginuma T, Gao J, Nagata K, Muroya R, Fei H, Nagano H, Chishaki S, Matsubara T, Kokabu S, Matsuo K, Kiyoshima T, Yoshioka I, Jimi E. p130Cas induces bone invasion by oral squamous cell carcinoma by regulating tumor epithelial-mesenchymal transition and cell proliferation. Carcinogenesis 2021; 41:1038-1048. [PMID: 31996896 DOI: 10.1093/carcin/bgaa007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 12/27/2019] [Accepted: 01/28/2020] [Indexed: 02/07/2023] Open
Abstract
Bone invasion is a critical factor in determining the prognosis of oral squamous cell carcinoma (OSCC) patients. Transforming growth factor β (TGF-β) is abundantly expressed in the bone matrix and is involved in the acquisition of aggressiveness by tumors. TGF-β is also important to cytoskeletal changes during tumor progression. In this study, we examined the relationship between TGF-β signaling and cytoskeletal changes during bone invasion by OSCC. Immunohistochemical staining of OSCC samples from five patients showed the expression of p130Cas (Crk-associated substrate) in the cytoplasm and phosphorylated Smad3 expression in the nucleus in OSCC cells. TGF-β1 induced the phosphorylation of Smad3 and p130Cas, as well as epithelial-mesenchymal transition (EMT) accompanied by the downregulation of the expression of E-cadherin, a marker of epithelial cells, and the upregulation of the expression of N-cadherin, or Snail, a marker of mesenchymal cells, in human HSC-2 cells and mouse squamous cell carcinome VII (SCCVII) cells. SB431542, a specific inhibitor of Smad2/3 signaling, abrogated the TGF-β1-induced phosphorylation of p130Cas and morphological changes. Silencing p130Cas using an short hairpin RNA (shRNA) or small interfering RNA in SCCVII cells suppressed TGF-β1-induced cell migration, invasion, EMT and matrix metalloproteinase-9 (MMP-9) production. Compared with control SCCVII cells, SCCVII cells with silenced p130Cas strongly suppressed zygomatic and mandibular destruction in vivo by reducing the number of osteoclasts, cell proliferation and MMP-9 production. Taken together, these results showed that the expression of TGF-β/p130Cas might be a new target for the treatment of OSCC bone invasion.
Collapse
Affiliation(s)
- Tatsuki Yaginuma
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Manazuru, Kokurakita-ku, Kitakyushu, Japan.,Division of Oral Medicine, Department of Oral and Maxillofacial Reconstructive Surgery, Kyushu Dental University, Manazuru, Kokurakita-ku, Kitakyushu, Japan.,Laboratory of Molecular and Cellular Biochemistry, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Jing Gao
- Laboratory of Molecular and Cellular Biochemistry, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Kengo Nagata
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Ryusuke Muroya
- Laboratory of Molecular and Cellular Biochemistry, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Huang Fei
- Laboratory of Molecular and Cellular Biochemistry, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Haruki Nagano
- Laboratory of Molecular and Cellular Biochemistry, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Sakura Chishaki
- Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Manazuru, Kokurakita-ku, Kitakyushu, Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Manazuru, Kokurakita-ku, Kitakyushu, Japan
| | - Kou Matsuo
- Division of Oral Pathology, Department of Health Improvement, Kyushu Dental University, Manazuru, Kokurakita-ku, Kitakyushu, Japan
| | - Tamotsu Kiyoshima
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Izumi Yoshioka
- Division of Oral Medicine, Department of Oral and Maxillofacial Reconstructive Surgery, Kyushu Dental University, Manazuru, Kokurakita-ku, Kitakyushu, Japan
| | - Eijiro Jimi
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Manazuru, Kokurakita-ku, Kitakyushu, Japan.,Laboratory of Molecular and Cellular Biochemistry, Maidashi, Higashi-ku, Fukuoka, Japan.,Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| |
Collapse
|
23
|
Gao YY, Ling ZY, Zhu YR, Shi C, Wang Y, Zhang XY, Zhang ZQ, Jiang Q, Chen MB, Yang S, Cao C. The histone acetyltransferase HBO1 functions as a novel oncogenic gene in osteosarcoma. Am J Cancer Res 2021; 11:4599-4615. [PMID: 33754016 PMCID: PMC7978299 DOI: 10.7150/thno.55655] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/06/2021] [Indexed: 02/06/2023] Open
Abstract
HBO1 (KAT7 or MYST2) is a histone acetyltransferase that acetylates H3 and H4 histones. Methods: HBO1 expression was tested in human OS tissues and cells. Genetic strategies, including shRNA, CRISPR/Cas9 and overexpression constructs, were applied to exogenously alter HBO1 expression in OS cells. The HBO1 inhibitor WM-3835 was utilized to block HBO1 activation. Results:HBO1 mRNA and protein expression is significantly elevated in OS tissues and cells. In established (MG63/U2OS lines) and primary human OS cells, shRNA-mediated HBO1 silencing and CRISPR/Cas9-induced HBO1 knockout were able to potently inhibit cell viability, growth, proliferation, as well as cell migration and invasion. Significant increase of apoptosis was detected in HBO1-silenced/knockout OS cells. Conversely, ectopic HBO1 overexpression promoted OS cell proliferation and migration. We identified ZNF384 (zinc finger protein 384) as a potential transcription factor of HBO1. Increased binding between ZNF384 and HBO1 promoter was detected in OS cell and tissues, whereas ZNF384 silencing via shRNA downregulated HBO1 and produced significant anti-OS cell activity. In vivo, intratumoral injection of HBO1 shRNA lentivirus silenced HBO1 and inhibited OS xenograft growth in mice. Furthermore, growth of HBO1-knockout OS xenografts was significantly slower than the control xenografts. WM-3835, a novel and high-specific small molecule HBO1 inhibitor, was able to potently suppressed OS cell proliferation and migration, and led to apoptosis activation. Furthermore, intraperitoneal injection of a single dose of WM-3835 potently inhibited OS xenograft growth in SCID mice. Conclusion: HBO1 overexpression promotes OS cell growth in vitro and in vivo.
Collapse
|
24
|
Characterising open chromatin in chick embryos identifies cis-regulatory elements important for paraxial mesoderm formation and axis extension. Nat Commun 2021; 12:1157. [PMID: 33608545 PMCID: PMC7895974 DOI: 10.1038/s41467-021-21426-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 01/25/2021] [Indexed: 01/31/2023] Open
Abstract
Somites arising from paraxial mesoderm are a hallmark of the segmented vertebrate body plan. They form sequentially during axis extension and generate musculoskeletal cell lineages. How paraxial mesoderm becomes regionalised along the axis and how this correlates with dynamic changes of chromatin accessibility and the transcriptome remains unknown. Here, we report a spatiotemporal series of ATAC-seq and RNA-seq along the chick embryonic axis. Footprint analysis shows differential coverage of binding sites for several key transcription factors, including CDX2, LEF1 and members of HOX clusters. Associating accessible chromatin with nearby expressed genes identifies cis-regulatory elements (CRE) for TCF15 and MEOX1. We determine their spatiotemporal activity and evolutionary conservation in Xenopus and human. Epigenome silencing of endogenous CREs disrupts TCF15 and MEOX1 gene expression and recapitulates phenotypic abnormalities of anterior-posterior axis extension. Our integrated approach allows dissection of paraxial mesoderm regulatory circuits in vivo and has implications for investigating gene regulatory networks.
Collapse
|
25
|
Guo AK, Itahana Y, Seshachalam VP, Chow HY, Ghosh S, Itahana K. Mutant TP53 interacts with BCAR1 to contribute to cancer cell invasion. Br J Cancer 2021; 124:299-312. [PMID: 33144694 PMCID: PMC7782524 DOI: 10.1038/s41416-020-01124-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 08/10/2020] [Accepted: 09/22/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Mutant TP53 interacts with other proteins to produce gain-of-function properties that contribute to cancer metastasis. However, the underlying mechanisms are still not fully understood. METHODS Using immunoprecipitation and proximity ligation assays, we evaluated breast cancer anti-estrogen resistance 1 (BCAR1) as a novel binding partner of TP53R273H, a TP53 mutant frequently found in human cancers. The biological functions of their binding were examined by the transwell invasion assay. Clinical outcome of patients was analysed based on TP53 status and BCAR1 expression using public database. RESULTS We discovered a novel interaction between TP53R273H and BCAR1. We found that BCAR1 translocates from the cytoplasm into the nucleus and binds to TP53R273H in a manner dependent on SRC family kinases (SFKs), which are known to enhance metastasis. The expression of full-length TP53R273H, but not the BCAR1 binding-deficient mutant TP53R273HΔ102-207, promoted cancer cell invasion. Furthermore, among the patients with mutant TP53, high BCAR1 expression was associated with a poorer prognosis. CONCLUSIONS The interaction between TP53R273H and BCAR1 plays an important role in enhancing cancer cell invasion. Thus, our study suggests a disruption of the TP53R273H-BCAR1 binding as a potential therapeutic approach for TP53R273H-harbouring cancer patients.
Collapse
Affiliation(s)
- Alvin Kunyao Guo
- Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Yoko Itahana
- Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | | | - Hui Ying Chow
- School of Applied Science, Temasek Polytechnic, 21 Tampines Avenue 1, Singapore, 529757, Singapore
| | - Sujoy Ghosh
- Centre for Computational Biology, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Koji Itahana
- Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.
| |
Collapse
|
26
|
Tong M, Suttapitugsakul S, Wu R. Effective Method for Accurate and Sensitive Quantitation of Rapid Changes of Newly Synthesized Proteins. Anal Chem 2020; 92:10048-10057. [PMID: 32531160 PMCID: PMC7425198 DOI: 10.1021/acs.analchem.0c01823] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein synthesis is quickly and tightly regulated in cells to adapt to the ever-changing extracellular and intracellular environment. Accurate quantitation of rapid protein synthesis changes can provide insights into protein functions and cellular activities, but it is very challenging to achieve because of the lack of effective analysis methods. Here, we developed an effective mass spectrometry-based method named quantitative O-propargyl-puromycin tagging (QOT) by integrating O-propargyl-puromycin (OPP) labeling, bioorthogonal chemistry, and multiplexed proteomics for global and quantitative analysis of rapid protein synthesis. The current method enables us to accurately quantitate rapid changes of newly synthesized proteins because, unlike amino acids and their analogs, OPP can be utilized by the ribosome immediately without being activated and conjugated to tRNA, and thus cell starvation or pretreatment is not required. This method was applied to quantitate rapid changes of protein synthesis in THP-1 macrophages treated with lipopolysaccharide (LPS). For 15-min labeling, >3000 proteins were quantitated, and the synthesis of 238 proteins was significantly altered, including transcription factors and cytokines. The results demonstrated that protein synthesis was modulated to facilitate protein secretion in macrophages in response to LPS. Considering the importance of protein synthesis, this method can be extensively applied to investigate rapid changes of protein synthesis in the biological and biomedical research fields.
Collapse
Affiliation(s)
- Ming Tong
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Suttipong Suttapitugsakul
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Ronghu Wu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
27
|
Kobayakawa M, Matsubara T, Mizokami A, Hiura F, Takakura N, Kokabu S, Matsuda M, Yasuda H, Nakamura I, Takei Y, Honda H, Hosokawa R, Jimi E. Kif1c regulates osteoclastic bone resorption as a downstream molecule of p130Cas. Cell Biochem Funct 2019; 38:300-308. [DOI: 10.1002/cbf.3476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 10/04/2019] [Accepted: 12/08/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Miki Kobayakawa
- Division of Molecular Signaling and Biochemistry, Department of Health ImprovementKyushu Dental University Kitakyushu Japan
- Division of Oral Reconstruction and Rehabilitation, Department of Oral Functional ReconstructionKyushu Dental University Kitakyushu Japan
- Laboratory of Molecular and Cellular BiochemistryFaculty of Dental Science, Kyushu University Fukuoka Japan
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health ImprovementKyushu Dental University Kitakyushu Japan
| | - Akiko Mizokami
- Oral Health/Brain Health/Total Health Research Center, Faculty of Dental ScienceKyushu University Fukuoka Japan
| | - Fumitaka Hiura
- Laboratory of Molecular and Cellular BiochemistryFaculty of Dental Science, Kyushu University Fukuoka Japan
| | - Nana Takakura
- Laboratory of Molecular and Cellular BiochemistryFaculty of Dental Science, Kyushu University Fukuoka Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health ImprovementKyushu Dental University Kitakyushu Japan
| | - Miho Matsuda
- Laboratory of Molecular and Cellular BiochemistryFaculty of Dental Science, Kyushu University Fukuoka Japan
| | - Hisataka Yasuda
- Nagahama Institute for Biochemical ScienceOriental Yeast Co., Ltd. Shiga Japan
| | - Ichiro Nakamura
- Faculty of Health and Medical ScienceTeikyo Heisei University 2‐51‐4 Higashi‐Ikebukuro Tokyo Japan
| | - Yosuke Takei
- Department of Anatomy and Neuroscience, Faculty of MedicineUniversity of Tsukuba Tsukuba Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory AnimalsTokyo Women's Medical University Tokyo Japan
| | - Ryuji Hosokawa
- Division of Oral Reconstruction and Rehabilitation, Department of Oral Functional ReconstructionKyushu Dental University Kitakyushu Japan
| | - Eijiro Jimi
- Division of Molecular Signaling and Biochemistry, Department of Health ImprovementKyushu Dental University Kitakyushu Japan
- Laboratory of Molecular and Cellular BiochemistryFaculty of Dental Science, Kyushu University Fukuoka Japan
- Oral Health/Brain Health/Total Health Research Center, Faculty of Dental ScienceKyushu University Fukuoka Japan
| |
Collapse
|
28
|
Yamamoto H, Hayakawa F, Yasuda T, Odaira K, Minamikawa Y, Tange N, Hirano D, Kojima Y, Morishita T, Tsuzuki S, Naoe T, Kiyoi H. ZNF384-fusion proteins have high affinity for the transcriptional coactivator EP300 and aberrant transcriptional activities. FEBS Lett 2019; 593:2151-2161. [PMID: 31234226 DOI: 10.1002/1873-3468.13506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 11/07/2022]
Abstract
Zinc-finger protein 384 (ZNF384) fusion (Z-fusion) genes have recently been identified as recurrent fusion genes in B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) and have been detected in 7-17% of Philadelphia chromosome-negative BCP-ALL cases. We selected SALL4 and ID2 as potential Z-fusion-specific transcriptional targets that might lead to the differentiation disorder of Z-fusion-positive ALL. The introduction of EP300-ZNF384 and SYNRG-ZNF384 induced the expression of these genes. Z-fusion proteins exhibited stronger transcriptional activities on the promoter or enhancer region of these genes than Wild-Z. Furthermore, GST pull-down assay revealed that Z-fusion proteins associated more strongly with EP300 than Wild-Z. Coexpression of EP300 specifically enhanced the transcriptional activities of Z-fusion proteins. We propose the increased EP300 binding of Z-fusion proteins as a mechanism for their increased transcriptional activities.
Collapse
Affiliation(s)
- Hideyuki Yamamoto
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| | - Fumihiko Hayakawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan.,Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Japan
| | - Takahiko Yasuda
- Clinical Research Center, Nagoya Medical Center, National Hospital Organization, Nagoya, Japan
| | - Koya Odaira
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Japan
| | - Yuka Minamikawa
- Department of Analytical Neurobiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Naoyuki Tange
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| | - Daiki Hirano
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| | - Yuki Kojima
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| | - Takanobu Morishita
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| | - Shinobu Tsuzuki
- Department of Biochemistry, School of Medicine, Aichi Medical University, Japan
| | - Tomoki Naoe
- Nagoya Medical Center, National Hospital Organization, Nagoya, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| |
Collapse
|
29
|
Overexpression of zinc finger protein 384 (ZNF 384), a poor prognostic predictor, promotes cell growth by upregulating the expression of Cyclin D1 in Hepatocellular carcinoma. Cell Death Dis 2019; 10:444. [PMID: 31168049 PMCID: PMC6551341 DOI: 10.1038/s41419-019-1681-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous, multigene-driven malignant tumor. ZNF384 is an overexpressed gene with a high frequency of alteration in HCC, but research on the function of ZNF384 in HCC is lacking. In this study, the expression level of ZNF384 in HCC was analyzed through immunohistochemical (IHC) staining, Western blot analysis and qRT-PCR. We also generated ZNF384 knockdown and knockout HCC cell lines using short hairpin RNA (shRNA) and CRISPR/Cas9 systems. MTS, colony formation, and 5-ethynyl-20-deoxyuridine (EdU) assays; flow cytometry; and a xenograft mouse model were used to evaluate the effects of ZNF384 on cell proliferation. Western blot analysis, a dual luciferase reporter assay and a ChIP assay were performed to explore the potential mechanism. We found that overexpression of ZNF384 in HCC and elevated expression of ZNF384 in HCC tissues was significantly correlated with tumor recurrence (P = 0.0097). Kaplan-Meier survival analysis revealed that high expression levels of ZNF384 were correlated with poor overall survival (P = 0.0386). Downregulation of ZNF384 expression suppressed HCC cell proliferation by inhibiting the expression of Cyclin D1. These findings suggest that ZNF384 tends to act as an oncogene in the development of HCC. ZNF384 promotes the proliferation of HCC cells by directly upregulating the expression of Cyclin D1 and might serve as a prognostic predictive factor for HCC patients.
Collapse
|
30
|
Georgakopoulos N, Diamantopoulos P, Micci F, Giannakopoulou N, Zervakis K, Dimitrakopoulou A, Viniou NA. An Adult Patient with Early Pre-B Acute Lymphoblastic Leukemia with t(12;17)(p13;q21)/ZNF384-TAF15. In Vivo 2018; 32:1241-1245. [PMID: 30150451 DOI: 10.21873/invivo.11371] [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: 04/23/2018] [Revised: 05/29/2018] [Accepted: 06/05/2018] [Indexed: 11/10/2022]
Abstract
This is a case report of a 46-year-old man diagnosed with early pre-B acute lymphoblastic leukemia (ALL), bearing the translocation t(12;17)(p13;q21) as the sole chromosomal abnormality. This is a rare chromosomal abnormality that has been reported in approximately 25 cases worldwide. FISH analysis revealed a rearrangement of ZNF384 (12p13) and TAF15 (17q12) genes, which is usually associated with a pre-B ALL phenotype with co-expression of the myeloid markers CD13 and/or CD33. ZNF384 encodes a zinc finger protein, which acts as a transcription factor, regulating the expression of several matrix metalloproteinases and TAF15 belongs to the FET (FUS, EWS, and TAF15) family, consisting of RNA and DNA-binding proteins. Unlike most of the cases where CD10 expression was absent or weak, in our case CD10 was highly expressed. The prognostic significance of ZNF384/TAF15 fusion is not very clear since several reports support a generally good prognosis, while others support a poor clinical outcome. Our patient was treated with the German multicenter ALL (GMALL) protocol for B-ALL, but experienced a fulminant gram-negative sepsis and eventually died during induction therapy.
Collapse
Affiliation(s)
| | - Panagiotis Diamantopoulos
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Francesca Micci
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Nefeli Giannakopoulou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Zervakis
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Aglaia Dimitrakopoulou
- Department of Immunology and Histocompatibility, Laikon General Hospital, Athens, Greece
| | - Nora-Athina Viniou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
31
|
Gemperle J, Hexnerová R, Lepšík M, Tesina P, Dibus M, Novotný M, Brábek J, Veverka V, Rosel D. Structural characterization of CAS SH3 domain selectivity and regulation reveals new CAS interaction partners. Sci Rep 2017; 7:8057. [PMID: 28808245 PMCID: PMC5556061 DOI: 10.1038/s41598-017-08303-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/06/2017] [Indexed: 12/22/2022] Open
Abstract
CAS is a docking protein downstream of the proto-oncogene Src with a role in invasion and metastasis of cancer cells. The CAS SH3 domain is indispensable for CAS-mediated signaling, but structural aspects of CAS SH3 ligand binding and regulation are not well understood. Here, we identified the consensus CAS SH3 binding motif and structurally characterized the CAS SH3 domain in complex with ligand. We revealed the requirement for an uncommon centrally localized lysine residue at position +2 of CAS SH3 ligands and two rather dissimilar optional anchoring residues, leucine and arginine, at position +5. We further expanded the knowledge of CAS SH3 ligand binding regulation by manipulating tyrosine 12 phosphorylation and confirmed the negative role of this phosphorylation on CAS SH3 ligand binding. Finally, by exploiting the newly identified binding requirements of the CAS SH3 domain, we predicted and experimentally verified two novel CAS SH3 binding partners, DOK7 and GLIS2.
Collapse
Affiliation(s)
- Jakub Gemperle
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, Czech Republic
| | - Rozálie Hexnerová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, Prague, Czech Republic
| | - Martin Lepšík
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, Prague, Czech Republic
| | - Petr Tesina
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, Prague, Czech Republic
| | - Michal Dibus
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, Czech Republic
| | - Marian Novotný
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, Czech Republic
| | - Václav Veverka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, Prague, Czech Republic.
| | - Daniel Rosel
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, Czech Republic.
| |
Collapse
|
32
|
The role of focal adhesion anchoring domains of CAS in mechanotransduction. Sci Rep 2017; 7:46233. [PMID: 28406229 PMCID: PMC5390273 DOI: 10.1038/srep46233] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/14/2017] [Indexed: 11/08/2022] Open
Abstract
CAS is a docking protein, which was shown to act as a mechanosensor in focal adhesions. The unique assembly of structural domains in CAS is important for its function as a mechanosensor. The tension within focal adhesions is transmitted to a stretchable substrate domain of CAS by focal adhesion-targeting of SH3 and CCH domain of CAS, which anchor the CAS protein in focal adhesions. Mechanistic models of the stretching biosensor propose equal roles for both anchoring domains. Using deletion mutants and domain replacements, we have analyzed the relative importance of the focal adhesion anchoring domains on CAS localization and dynamics in focal adhesions as well as on CAS-mediated mechanotransduction. We confirmed the predicted prerequisite of the focal adhesion targeting for CAS-dependent mechanosensing and unraveled the critical importance of CAS SH3 domain in mechanosensing. We further show that CAS localizes to the force transduction layer of focal adhesions and that mechanical stress stabilizes CAS in focal adhesions.
Collapse
|
33
|
Yaguchi A, Ishibashi T, Terada K, Ueno-Yokohata H, Saito Y, Fujimura J, Shimizu T, Ohki K, Manabe A, Kiyokawa N. EP300-ZNF384 fusion gene product up-regulates GATA3 gene expression and induces hematopoietic stem cell gene expression signature in B-cell precursor acute lymphoblastic leukemia cells. Int J Hematol 2017; 106:269-281. [PMID: 28378055 DOI: 10.1007/s12185-017-2220-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/20/2017] [Accepted: 03/21/2017] [Indexed: 01/08/2023]
Abstract
ZNF384-related fusion genes are associated with a distinct subgroup of B-cell precursor acute lymphoblastic leukemias in childhood, with a frequency of approximately 3-4%. We previously identified a novel EP300-ZNF384 fusion gene. Patients with the ZNF384-related fusion gene exhibit a hematopoietic stem cell (HSC) gene expression signature and characteristic immunophenotype with negative or low expression of CD10 and aberrant expression of myeloid antigens, such as CD33 and CD13. However, the molecular basis of this pathogenesis remains completely unknown. In the present study, we examined the biological effects of EP300-ZNF384 expression induced by retrovirus-mediated gene transduction in an REH B-cell precursor acute lymphoblastic leukemia cell line, and observed the acquisition of the HSC gene expression signature and an up-regulation of GATA3 gene expression, as assessed by microarray analysis. In contrast, the gene expression profile induced by wild-type ZNF384 in REH cells was significantly different from that by EP300-ZNF384 expression. Together with the results of reporter assays, which revealed the enhancement of GATA3-promoter activity by EP300-ZNF384 expression, these findings suggest that EP300-ZNF384 mediates GATA3 gene expression and may be involved in the acquisition of the HSC gene expression signature and characteristic immunophenotype in B-cell precursor acute lymphoblastic leukemia cells.
Collapse
Affiliation(s)
- Akinori Yaguchi
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Takeshi Ishibashi
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kazuki Terada
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Hitomi Ueno-Yokohata
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Yuya Saito
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Junya Fujimura
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kentaro Ohki
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Atsushi Manabe
- Department of Pediatrics, St. Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| | - Nobutaka Kiyokawa
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan.
| |
Collapse
|
34
|
Shago M, Abla O, Hitzler J, Weitzman S, Abdelhaleem M. Frequency and outcome of pediatric acute lymphoblastic leukemia with ZNF384 gene rearrangements including a novel translocation resulting in an ARID1B/ZNF384 gene fusion. Pediatr Blood Cancer 2016; 63:1915-21. [PMID: 27392123 DOI: 10.1002/pbc.26116] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/17/2016] [Accepted: 05/31/2016] [Indexed: 11/11/2022]
Abstract
BACKGROUND ZNF384 gene rearrangements with multiple partner genes are recurrent in acute leukemia and are most often associated with a precursor B cell immunophenotype. The overall incidence of this genetic category of leukemia is uncertain. PROCEDURE Patients with ZNF384 gene rearrangements from a cohort of 240 precursor B cell acute lymphoblastic leukemia (ALL) pediatric patients over a 3.5-year time period were characterized with detailed cytogenetic, FISH, genomic, and clinical analyses. RESULTS Seven of the 240 patients were identified to have ZNF384 gene rearrangements including partner genes TCF3 (four patients), EWSR1 (one patient), EP300 (one patient), and the novel gene partner ARID1B (one patient). The translocations were confirmed by FISH analysis and with RNA sequencing for the EP300 and ARID1B partner genes. Genomic microarray analysis showed an average of 2.7 copy number alterations in each case with no evidence of imbalance at the translocation breakpoints. Six of the patients with ZNF384 gene rearrangements had precursor B cell ALL with a CD10- immunophenotype and myeloid-associated antigens. One of the patients also had myeloperoxidase expression and was diagnosed as mixed phenotype B/myeloid acute leukemia. None of the patients have relapsed with event-free survival ranging from 6 years 2 months to 9 years 2 months. CONCLUSIONS This study suggests that the frequency of ZNF384 gene rearrangement in pediatric precursor B cell ALL is approximately 3%. The ARID1B gene, commonly mutated in multiple types of cancer, was identified as an additional ZNF384 gene fusion partner.
Collapse
Affiliation(s)
- Mary Shago
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| | - Oussama Abla
- Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Johann Hitzler
- Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Sheila Weitzman
- Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Mohamed Abdelhaleem
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Young SK, Shao Y, Bidwell JP, Wek RC. Nuclear Matrix Protein 4 Is a Novel Regulator of Ribosome Biogenesis and Controls the Unfolded Protein Response via Repression of Gadd34 Expression. J Biol Chem 2016; 291:13780-8. [PMID: 27129771 DOI: 10.1074/jbc.m116.729830] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Indexed: 12/24/2022] Open
Abstract
The unfolded protein response (UPR) maintains protein homeostasis by governing the processing capacity of the endoplasmic reticulum (ER) to manage ER client loads; however, key regulators within the UPR remain to be identified. Activation of the UPR sensor PERK (EIFAK3/PEK) results in the phosphorylation of the α subunit of eIF2 (eIF2α-P), which represses translation initiation and reduces influx of newly synthesized proteins into the overloaded ER. As part of this adaptive response, eIF2α-P also induces a feedback mechanism through enhanced transcriptional and translational expression of Gadd34 (Ppp1r15A),which targets type 1 protein phosphatase for dephosphorylation of eIF2α-P to restore protein synthesis. Here we describe a novel mechanism by which Gadd34 expression is regulated through the activity of the zinc finger transcription factor NMP4 (ZNF384, CIZ). NMP4 functions to suppress bone anabolism, and we suggest that this occurs due to decreased protein synthesis of factors involved in bone formation through NMP4-mediated dampening of Gadd34 and c-Myc expression. Loss of Nmp4 resulted in an increase in c-Myc and Gadd34 expression that facilitated enhanced ribosome biogenesis and global protein synthesis. Importantly, protein synthesis was sustained during pharmacological induction of the UPR through a mechanism suggested to involve GADD34-mediated dephosphorylation of eIF2α-P. Sustained protein synthesis sensitized cells to pharmacological induction of the UPR, and the observed decrease in cell viability was restored upon inhibition of GADD34 activity. We conclude that NMP4 is a key regulator of ribosome biogenesis and the UPR, which together play a central role in determining cell viability during endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Sara K Young
- From the Department of Biochemistry and Molecular Biology
| | - Yu Shao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202-5126
| | | | - Ronald C Wek
- From the Department of Biochemistry and Molecular Biology,
| |
Collapse
|
36
|
Rump K, Siffert W, Peters J, Adamzik M. The Transcription Factor NMP4 Binds to the AQP5 Promoter and Is a Novel Transcriptional Regulator of the AQP5 Gene. DNA Cell Biol 2016; 35:322-7. [PMID: 27058007 DOI: 10.1089/dna.2015.3110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aquaporin 5 (AQP5) is a water channel that regulates water transport, cell migration, and proliferation. Therefore, knowledge of its genetic regulation could be relevant to study these mechanisms. The AQP5 promoter region containing the AQP5-1364 A/C single-nucleotide polymorphism (SNP) might be an important regulatory region because the SNP is associated with the etiopathology of several diseases. The aim of this study was to identify a transcription factor that binds to this AQP5 promoter region and to investigate its potential impact upon AQP5 expression. In silico analysis revealed nuclear matrix protein 4 (NMP4) as a putative candidate. Electrophoretic mobility shift assays showed specific binding of NMP4 to the AQP5 promoter region of nt -1370 to nt -1329. Overexpression of NMP4 increased AQP5 promoter activity of the analyzed promoter constructs from nt -469 to nt -1979. Furthermore, an additional NMP4 binding site at position nt -592/nt -602 of the AQP5 promoter was identified. NMP4 overexpression increased AQP5 mRNA expression by 2.5-fold in HEK293 cells. Summarizing, we identified NMP4 as a novel transcriptional regulator of AQP5 expression, which binds to two AQP5 promoter regions. Both regions appear to impact AQP5 expression significantly.
Collapse
Affiliation(s)
- Katharina Rump
- 1 Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum-Langendreer der Ruhr-Universität Bochum , Bochum, Germany .,2 Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen , Essen, Germany .,3 Institut für Pharmakogenetik, Universität Duisburg-Essen, Universitätsklinikum Essen , Essen, Germany
| | - Winfried Siffert
- 3 Institut für Pharmakogenetik, Universität Duisburg-Essen, Universitätsklinikum Essen , Essen, Germany
| | - Jürgen Peters
- 2 Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen , Essen, Germany
| | - Michael Adamzik
- 1 Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum-Langendreer der Ruhr-Universität Bochum , Bochum, Germany .,2 Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen , Essen, Germany
| |
Collapse
|
37
|
Nakamoto T, Izu Y, Kawasaki M, Notomi T, Hayata T, Noda M, Ezura Y. Mice Deficient in CIZ/NMP4 Develop an Attenuated Form of K/BxN-Serum Induced Arthritis. J Cell Biochem 2015; 117:970-7. [PMID: 26378628 DOI: 10.1002/jcb.25382] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 09/15/2015] [Indexed: 01/08/2023]
Abstract
CIZ/NMP4 (Cas interacting zinc finger protein, Nmp4, Zfp384) is a transcription factor that is known to regulate matrix related-proteins. To explore the possible pathophysiological role of CIZ/NMP4 in arthritis, we examined CIZ/NMP4 expression in articular cartilage in arthritis model. CIZ/NMP4 was expressed in the articular chondrocytes of mice at low levels while its expression was enhanced when arthritis was induced. Arthritis induction increased clinical score in wild type mice. In contrast, CIZ/NMP4 deficiency suppressed such rise in the levels of arthritis score and swelling of soft tissue. CIZ/NMP4 deficiency also reduced invasion of inflammatory cells in joint tissue. Quantitative PCR analyses of mRNA from joints revealed that arthritis-induced increase in expressions of IL-1β was suppressed by CIZ/NMP4 deficiency. CIZ/NMP4 bound to IL-1β promoter and activated its transcription. The increase in CIZ/NMP4 in arthritis was also associated with enhancement in bone resorption and cartilage matrix degradation. In fact, RANKL, a signaling molecule prerequisite for osteoclastogenesis and, MMP-3, a clinical marker for arthritis were increased in joints upon arthritis induction. In contrast, CIZ/NMP4 deficiency suppressed the arthritis-induced increase in bone resorption, expression of RANKL and MMP-3 mRNA. Thus, CIZ/NMP4 plays a role in the development of arthritis at least in part through regulation of key molecules related to the arthritis.
Collapse
Affiliation(s)
- Tetsuya Nakamoto
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Yayoi Izu
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Makiri Kawasaki
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Takuya Notomi
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Tadayoshi Hayata
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Masaki Noda
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Yoichi Ezura
- Department of Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| |
Collapse
|
38
|
Rodríguez-Fernández JL, de Lacoba MG. Plasma membrane-associated superstructure: Have we overlooked a new type of organelle in eukaryotic cells? J Theor Biol 2015; 380:346-58. [PMID: 26066286 DOI: 10.1016/j.jtbi.2015.05.029] [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: 10/14/2014] [Revised: 05/22/2015] [Accepted: 05/25/2015] [Indexed: 10/23/2022]
Abstract
A variety of intriguing plasma membrane-associated regions, including focal adhesions, adherens junctions, tight junctions, immunological synapses, neuromuscular junctions and the primary cilia, among many others, have been described in eukaryotic cells. Emphasizing their importance, alteration in their molecular structures induces or correlates with different pathologies. These regions display surface proteins connected to intracellular molecules, including cytoskeletal component, which maintain their cytoarchitecture, and signalling proteins, which regulate their organization and functions. Based on the molecular similarities and other common features observed, we suggest that, despite differences in external appearances, all these regions are just the same superstructure that appears in different locations and cells. We hypothesize that this superstructure represents an overlooked new type of organelle that we call plasma membrane-associated superstructure (PMAS). Therefore, we suggest that eukaryotic cells include classical organelles (e.g. mitochondria, Golgi and others) and also PMAS. We speculate that this new type of organelle might be an innovation associated to the emergence of eukaryotes. Finally we discuss the implications of the hypothesis proposed.
Collapse
Affiliation(s)
- José Luis Rodríguez-Fernández
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, 9, Madrid 28040, Spain.
| | - Mario García de Lacoba
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, 9, Madrid 28040, Spain
| |
Collapse
|
39
|
Childress P, Stayrook KR, Alvarez MB, Wang Z, Shao Y, Hernandez-Buquer S, Mack JK, Grese ZR, He Y, Horan D, Pavalko FM, Warden SJ, Robling AG, Yang FC, Allen MR, Krishnan V, Liu Y, Bidwell JP. Genome-Wide Mapping and Interrogation of the Nmp4 Antianabolic Bone Axis. Mol Endocrinol 2015; 29:1269-85. [PMID: 26244796 DOI: 10.1210/me.2014-1406] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PTH is an osteoanabolic for treating osteoporosis but its potency wanes. Disabling the transcription factor nuclear matrix protein 4 (Nmp4) in healthy, ovary-intact mice enhances bone response to PTH and bone morphogenetic protein 2 and protects from unloading-induced osteopenia. These Nmp4(-/-) mice exhibit expanded bone marrow populations of osteoprogenitors and supporting CD8(+) T cells. To determine whether the Nmp4(-/-) phenotype persists in an osteoporosis model we compared PTH response in ovariectomized (ovx) wild-type (WT) and Nmp4(-/-) mice. To identify potential Nmp4 target genes, we performed bioinformatic/pathway profiling on Nmp4 chromatin immunoprecipitation sequencing (ChIP-seq) data. Mice (12 w) were ovx or sham operated 4 weeks before the initiation of PTH therapy. Skeletal phenotype analysis included microcomputed tomography, histomorphometry, serum profiles, fluorescence-activated cell sorting and the growth/mineralization of cultured WT and Nmp4(-/-) bone marrow mesenchymal stem progenitor cells (MSPCs). ChIP-seq data were derived using MC3T3-E1 preosteoblasts, murine embryonic stem cells, and 2 blood cell lines. Ovx Nmp4(-/-) mice exhibited an improved response to PTH coupled with elevated numbers of osteoprogenitors and CD8(+) T cells, but were not protected from ovx-induced bone loss. Cultured Nmp4(-/-) MSPCs displayed enhanced proliferation and accelerated mineralization. ChIP-seq/gene ontology analyses identified target genes likely under Nmp4 control as enriched for negative regulators of biosynthetic processes. Interrogation of mRNA transcripts in nondifferentiating and osteogenic differentiating WT and Nmp4(-/-) MSPCs was performed on 90 Nmp4 target genes and differentiation markers. These data suggest that Nmp4 suppresses bone anabolism, in part, by regulating IGF-binding protein expression. Changes in Nmp4 status may lead to improvements in osteoprogenitor response to therapeutic cues.
Collapse
Affiliation(s)
- Paul Childress
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Keith R Stayrook
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Marta B Alvarez
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Zhiping Wang
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Yu Shao
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Selene Hernandez-Buquer
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Justin K Mack
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Zachary R Grese
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Yongzheng He
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Daniel Horan
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Fredrick M Pavalko
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Stuart J Warden
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Alexander G Robling
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Feng-Chun Yang
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Matthew R Allen
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Venkatesh Krishnan
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Yunlong Liu
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| | - Joseph P Bidwell
- Department of Anatomy and Cell Biology (P.C., S.H.-B., D.H., A.G.R., M.R.A., J.P.B.), Indiana University School of Medicine, Indianapolis, Indiana 46202; Lilly Research Laboratories (K.R.S., J.K.M., Z.R.G., V.K.), Eli Lilly and Company, Indianapolis, Indiana 46202; Orthopaedic Surgery (M.B.A.), Indiana University School of Medicine; Department of Medical and Molecular Genetics (Z.W., Y.S., Y.L., J.P.B.), Indiana University School of Medicine; Center for Computational Biology and Bioinformatics (Z.W., Y.L.), Indiana University School of Medicine; Department of Pediatrics (Y.H., F.-C.Y.), Indiana University School of Medicine; Herman B Wells Center for Pediatric Research (Y.H., F.-C.Y.); Cellular and Integrative Physiology (F.M.P.); Center for Translational Musculoskeletal Research (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University; and Department of Physical Therapy (S.J.W.), School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, Indiana 46202
| |
Collapse
|
40
|
Deneka A, Korobeynikov V, Golemis EA. Embryonal Fyn-associated substrate (EFS) and CASS4: The lesser-known CAS protein family members. Gene 2015; 570:25-35. [PMID: 26119091 DOI: 10.1016/j.gene.2015.06.062] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 06/23/2015] [Indexed: 01/15/2023]
Abstract
The CAS (Crk-associated substrate) adaptor protein family consists of four members: CASS1/BCAR1/p130Cas, CASS2/NEDD9/HEF1/Cas-L, CASS3/EFS/Sin and CASS4/HEPL. While CAS proteins lack enzymatic activity, they contain specific recognition and binding sites for assembly of larger signaling complexes that are essential for cell proliferation, survival, migration, and other processes. All family members are intermediates in integrin-dependent signaling pathways mediated at focal adhesions, and associate with FAK and SRC family kinases to activate downstream effectors regulating the actin cytoskeleton. Most studies of CAS proteins to date have been focused on the first two members, BCAR1 and NEDD9, with altered expression of these proteins now appreciated as influencing disease development and prognosis for cancer and other serious pathological conditions. For these family members, additional mechanisms of action have been defined in receptor tyrosine kinase (RTK) signaling, estrogen receptor signaling or cell cycle progression, involving discrete partner proteins such as SHC, NSP proteins, or AURKA. By contrast, EFS and CASS4 have been less studied, although structure-function analyses indicate they conserve many elements with the better-known family members. Intriguingly, a number of recent studies have implicated these proteins in immune system function, and the pathogenesis of developmental disorders, autoimmune disorders including Crohn's disease, Alzheimer's disease, cancer and other diseases. In this review, we summarize the current understanding of EFS and CASS4 protein function in the context of the larger CAS family group.
Collapse
Affiliation(s)
- Alexander Deneka
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, United States; Kazan Federal University, 420000, Kazan, Russian Federation
| | - Vladislav Korobeynikov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, United States; Novosibirsk State University, Medical Department, 630090, Novosibirsk, Russian Federation
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, United States.
| |
Collapse
|
41
|
Identification of APOBEC3B promoter elements responsible for activation by human papillomavirus type 16 E6. Biochem Biophys Res Commun 2015; 460:555-60. [PMID: 25800874 DOI: 10.1016/j.bbrc.2015.03.068] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/12/2015] [Indexed: 12/13/2022]
Abstract
Recent cancer genomics studies have identified mutation patterns characteristic of APOBEC3B (A3B) in multiple cancers, including cervical cancer, which is caused by human papillomavirus (HPV) infection. A3B expression is upregulated by HPV E6/E7 oncoproteins, implying a crucial role for A3B upregulation in HPV-induced carcinogenesis. Here, we explored the molecular mechanisms underlying the activation of the A3B promoter by E6. Luciferase reporter assays with a series of deleted fragments of the human A3B promoter in normal immortalized human keratinocytes (NIKS) identified two functional regions in the promoter: the distal region (from -200 to -51), which is required for basal promoter activity, and the proximal region (from +1 to +45), which exerts an inhibitory effect on gene expression. Each promoter region was found to contain an E6-responsive element(s). Disruption of an AT-rich motif located between +10 and +16 abrogated the proximal-region-mediated activation of the A3B promoter by E6. DNA pull-down assays revealed that a cellular zinc-finger protein, ZNF384, binds to the AT-rich motif in the A3B promoter, and chromatin immunoprecipitation assays confirmed that ZNF384 binds to the A3B promoter in cells. ZNF384 knockdown reduced the A3B mRNA levels in NIKS expressing E6, but not in the parental NIKS, indicating that ZNF384 contributes to A3B upregulation by E6, but not to basal A3B expression. The exogenous expression of ZNF384 led to the activation of the A3B promoter in NIKS. Collectively, these results indicate that E6 activates the A3B promoter through the distal and proximal regions, and that ZNF384 is required for the proximal-region-mediated activation of A3B.
Collapse
|
42
|
Janoštiak R, Pataki AC, Brábek J, Rösel D. Mechanosensors in integrin signaling: The emerging role of p130Cas. Eur J Cell Biol 2014; 93:445-54. [DOI: 10.1016/j.ejcb.2014.07.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/11/2014] [Accepted: 07/01/2014] [Indexed: 12/17/2022] Open
|
43
|
Johnson JL. Emerging regulators of vascular smooth muscle cell function in the development and progression of atherosclerosis. Cardiovasc Res 2014; 103:452-60. [PMID: 25053639 DOI: 10.1093/cvr/cvu171] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
After a period of relative senescence in the field of vascular smooth muscle cell (VSMC) research with particular regards to atherosclerosis, the last few years has witnessed a resurgence, with extensive research re-assessing potential molecular mechanisms and pathways that modulate VSMC behaviour within the atherosclerotic-prone vessel wall and the atherosclerotic plaque itself. Attention has focussed on the pathological contribution of VSMC in plaque calcification; systemic and local mediators such as inflammatory molecules and lipoproteins; autocrine and paracrine regulators which affect cell-cell and cell to matrix contacts alongside cytoskeletal changes. In this brief focused review, recent insights that have been gained into how a myriad of recently identified factors can influence the pathological behaviour of VSMC and their subsequent contribution to atherosclerotic plaque development and progression has been discussed. An overriding theme is the mechanisms involved in the alterations of VSMC function during atherosclerosis.
Collapse
Affiliation(s)
- Jason Lee Johnson
- Laboratory of Cardiovascular Pathology, School of Clinical Sciences, University of Bristol, Research Floor Level Seven, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| |
Collapse
|
44
|
CAS directly interacts with vinculin to control mechanosensing and focal adhesion dynamics. Cell Mol Life Sci 2013; 71:727-44. [PMID: 23974298 PMCID: PMC3901934 DOI: 10.1007/s00018-013-1450-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/19/2013] [Accepted: 08/05/2013] [Indexed: 12/31/2022]
Abstract
Focal adhesions are cellular structures through which both mechanical forces and regulatory signals are transmitted. Two focal adhesion-associated proteins, Crk-associated substrate (CAS) and vinculin, were both independently shown to be crucial for the ability of cells to transmit mechanical forces and to regulate cytoskeletal tension. Here, we identify a novel, direct binding interaction between CAS and vinculin. This interaction is mediated by the CAS SRC homology 3 domain and a proline-rich sequence in the hinge region of vinculin. We show that CAS localization in focal adhesions is partially dependent on vinculin, and that CAS–vinculin coupling is required for stretch-induced activation of CAS at the Y410 phosphorylation site. Moreover, CAS–vinculin binding significantly affects the dynamics of CAS and vinculin within focal adhesions as well as the size of focal adhesions. Finally, disruption of CAS binding to vinculin reduces cell stiffness and traction force generation. Taken together, these findings strongly implicate a crucial role of CAS–vinculin interaction in mechanosensing and focal adhesion dynamics.
Collapse
|
45
|
Barrett A, Pellet-Many C, Zachary IC, Evans IM, Frankel P. p130Cas: a key signalling node in health and disease. Cell Signal 2012; 25:766-77. [PMID: 23277200 DOI: 10.1016/j.cellsig.2012.12.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 12/21/2012] [Indexed: 01/08/2023]
Abstract
p130Cas/breast cancer anti-oestrogen resistance 1 (BCAR1) is a member of the Cas (Crk-associated substrate) family of adaptor proteins, which have emerged as key signalling nodes capable of interactions with multiple proteins, with important regulatory roles in normal and pathological cell function. The Cas family of proteins is characterised by the presence of multiple conserved motifs for protein-protein interactions, and by extensive tyrosine and serine phosphorylations. Recent studies show that p130Cas contributes to migration, cell cycle control and apoptosis. p130Cas is essential during early embryogenesis, with a critical role in cardiovascular development. Furthermore, p130Cas has been reported to be involved in the development and progression of several human cancers. p130Cas is able to perform roles in multiple processes due to its capacity to regulate a diverse array of signalling pathways, transducing signals from growth factor receptor tyrosine kinases, non-receptor tyrosine kinases, and integrins. In this review we summarise the current understanding of the structure, function, and regulation of p130Cas, and discuss the importance of p130Cas in both physiological and pathophysiological settings, with a focus on the cardiovascular system and cancer.
Collapse
Affiliation(s)
- Angela Barrett
- Centre for Cardiovascular Biology and Medicine, Division of Medicine, University College London, London WC1E 6JJ, United Kingdom.
| | | | | | | | | |
Collapse
|
46
|
Bidwell JP, Childress P, Alvarez MB, Hood M, He Y, Pavalko FM, Kacena MA, Yang FC. Nmp4/CIZ closes the parathyroid hormone anabolic window. Crit Rev Eukaryot Gene Expr 2012; 22:205-18. [PMID: 23140162 DOI: 10.1615/critreveukargeneexpr.v22.i3.40] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic degenerative diseases are increasing with the aging U.S. population. One consequence of this phenomenon is the need for long-term osteoporosis therapies. Parathyroid hormone (PTH), the only FDA-approved treatment that adds bone to the aged skeleton, loses its potency within two years of initial treatment but the mechanism regulating its limited "anabolic window" is unknown. We have discovered that disabling the nucleocytoplasmic shuttling transcription factor nuclear matrix protein 4/cas interacting zinc finger protein (Nmp4/CIZ) in mice extends the PTH bone-forming capacity. Nmp4 was discovered during our search for nuclear matrix transcription factors that couple this hormone's impact on osteoblast cytoskeletal and nuclear organization with its anabolic capacity. CIZ was independently discovered as a protein that associates with the focal adhesion-associated mechanosensor p130Cas. The Nmp4/CIZ-knockout (KO) skeletal phenotype exhibits a modestly enhanced bone mineral density but manifests an exaggerated response to both PTH and to BMP2 and is resistant to disuse-induced bone loss. The cellular basis of the global Nmp4/CIZ-KO skeletal phenotype remains to be elucidated but may involve an expansion of the bone marrow osteoprogenitor population along with modestly enhanced osteoblast and osteoclast activities supporting anabolic bone turnover. As a shuttling Cys(2)His(2) zinc finger protein, Nmp4/CIZ acts as a repressive transcription factor perhaps associated with epigenetic remodeling complexes, but the functional significance of its interaction with p130Cas is not known. Despite numerous remaining questions, Nmp4/CIZ provides insights into how the anabolic window is regulated, and itself may provide an adjuvant therapy target for the treatment of osteoporosis by extending PTH anabolic efficacy.
Collapse
Affiliation(s)
- Joseph P Bidwell
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Rakar J, Lönnqvist S, Sommar P, Junker J, Kratz G. Interpreted gene expression of human dermal fibroblasts after adipo-, chondro- and osteogenic phenotype shifts. Differentiation 2012; 84:305-13. [DOI: 10.1016/j.diff.2012.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 07/30/2012] [Accepted: 08/19/2012] [Indexed: 11/27/2022]
|
48
|
Sakuma T, Nakamoto T, Hemmi H, Kitazawa S, Kitazawa R, Notomi T, Hayata T, Ezura Y, Amagasa T, Noda M. CIZ/NMP4 is expressed in B16 melanoma and forms a positive feedback loop with RANKL to promote migration of the melanoma cells. J Cell Physiol 2012; 227:2807-12. [PMID: 22307584 DOI: 10.1002/jcp.24066] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor metastasis to bone is a serious pathological situation that causes severe pain, and deterioration in locomoter function. However, the mechanisms underlying tumor metastasis is still incompletely understood. CIZ/NMP4 is a nucleocytoplasmic shuttling protein and its roles in tumor cells have not been known. We, therefore, hypothesized the role of CIZ/NMP4 in B16 melanoma cells that metastasize to bone. CIZ/NMP4 is expressed in B16 cells. The CIZ/NMP4 expression levels are correlated to the metastatic activity in divergent types of melanoma cells. Overexpression of CIZ/NMP4 increased B16 cell migration in Trans-well assay. Conversely, siRNA-based knockdown of CIZ/NMP4 suppressed migratory activity of these cells. As RANKL promotes metastasis of tumor cells in bone, we tested its effect on CIZ in melanoma cells. RANKL treatment enhanced CIZ/NMP4 expression. This increase of CIZ by RANKL promoted migration. Conversely, we identified CIZ/NMP4 binding site in the promoter of RANKL. Furthermore, luciferase assay indicated that CIZ/NMP4 overexpression enhanced RANKL promoter activities, revealing a positive feedback loop of CIZ/NMP4 and RANKL in melanoma. These observations indicate that CIZ/NMP4 is critical regulator of metastasis of melanoma cells.
Collapse
Affiliation(s)
- Tomomi Sakuma
- Molecular Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Jadwin JA, Ogiue-Ikeda M, Machida K. The application of modular protein domains in proteomics. FEBS Lett 2012; 586:2586-96. [PMID: 22710164 DOI: 10.1016/j.febslet.2012.04.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Revised: 04/13/2012] [Accepted: 04/13/2012] [Indexed: 11/19/2022]
Abstract
The ability of modular protein domains to independently fold and bind short peptide ligands both in vivo and in vitro has allowed a significant number of protein-protein interaction studies to take advantage of them as affinity and detection reagents. Here, we refer to modular domain based proteomics as "domainomics" to draw attention to the potential of using domains and their motifs as tools in proteomics. In this review we describe core concepts of domainomics, established and emerging technologies, and recent studies by functional category. Accumulation of domain-motif binding data should ultimately provide the foundation for domain-specific interactomes, which will likely reveal the underlying substructure of protein networks as well as the selectivity and plasticity of signal transduction.
Collapse
Affiliation(s)
- Joshua A Jadwin
- Department of Genetics and Developmental Biology, Raymond and Beverly Sackler Laboratory of Genetics and Molecular Medicine, University of Connecticut Health Center, 400 Farmington Avenue, Farmington, CT 06030, USA
| | | | | |
Collapse
|
50
|
Alvarez MB, Childress P, Philip BK, Gerard-O'Riley R, Hanlon M, Herbert BS, Robling AG, Pavalko FM, Bidwell JP. Immortalization and characterization of osteoblast cell lines generated from wild-type and Nmp4-null mouse bone marrow stromal cells using murine telomerase reverse transcriptase (mTERT). J Cell Physiol 2012; 227:1873-82. [PMID: 21732358 DOI: 10.1002/jcp.22915] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Intermittent parathyroid hormone (PTH) adds new bone to the osteoporotic skeleton; the transcription factor Nmp4/CIZ represses PTH-induced bone formation in mice and as a consequence is a potential drug target for improving hormone clinical efficacy. To explore the impact of Nmp4/CIZ on osteoblast phenotype, we immortalized bone marrow stromal cells from wildtype (WT) and Nmp4-knockout (KO) mice using murine telomerase reverse transcriptase. Clonal lines were initially chosen based on their positive staining for alkaline phosphatase and capacity for mineralization. Disabling Nmp4/CIZ had no gross impact on osteoblast phenotype development. WT and KO clones exhibited identical sustained growth, reduced population doubling times, extended maintenance of the mature osteoblast phenotype, and competency for differentiating toward the osteoblast and adipocyte lineages. Additional screening of the immortalized cells for PTH-responsiveness permitted further studies with single WT and KO clones. We recently demonstrated that PTH-induced c-fos femoral mRNA expression is enhanced in Nmp4-KO mice and in the present study we observed that hormone stimulated either an equivalent or modestly enhanced increase in c-fos mRNA expression in both primary null and KO clone cells depending on PTH concentration. The null primary osteoblasts and KO clone cells exhibited a transiently enhanced response to bone morphogenetic protein 2 (BMP2). The clones exhibited lower and higher expressions of the PTH receptor (Pthr1) and the BMP2 receptor (Bmpr1a, Alk3), respectively, as compared to primary cells. These immortalized cell lines will provide a valuable tool for disentangling the complex functional roles underlying Nmp4/CIZ regulation of bone anabolism.
Collapse
Affiliation(s)
- Marta B Alvarez
- Department of Anatomy & Cell Biology, Indiana University School of Medicine (IUSM), Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|