1
|
Li Y, Yang Z, Xu M, Guan H, Wu Z, Li S. CALD1 inhibits invasion of human ovarian cancer cells by affecting cytoskeletal structure and the number of focal adhesion. Transl Cancer Res 2025; 14:1323-1335. [PMID: 40104711 PMCID: PMC11912064 DOI: 10.21037/tcr-24-1375] [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: 08/06/2024] [Accepted: 12/04/2024] [Indexed: 03/20/2025]
Abstract
Background Ovarian cancer (OV) is associated with the highest mortality rate among gynecological cancers, largely due to late diagnosis and chemoresistance. The identification of novel diagnostic markers and therapeutic targets is crucial. Caldesmon 1 (CALD1), a cytoskeleton-regulating protein, has been implicated in various cancers. This study aims to investigate the expression and functional significance of CALD1 in OV, focusing on its potential impact on cell invasion and metastasis. Methods We analyzed CALD1 expression using The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases, along with tissue microarray immunohistochemistry (IHC). Drug sensitivity analysis was performed using the 'oncopredict' R package. A CALD1 gene network was constructed, followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. SK-OV-3 cell lines with stable CALD1 knockdown were established and verified by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot (WB). We then assessed cell invasiveness using Transwell assays and visualized cytoskeletal changes through immunofluorescence staining of F-actin and Vinculin. Results The expression of CALD1 was significantly reduced in OV tissues compared to normal tissues. Patients with high and low expression levels of CALD1 showed significant differences in their response to chemotherapeutic drugs. CALD1 and its related genes were found to play an essential role in regulating cytoskeleton organization, focal adhesion formation, and cell movement processes. CALD1 knockdown cells exhibited a significant reduction in F-actin stress fibers, a loose cytoskeleton structure, decreased Vinculin expression, and enhanced migration ability. Conclusions Attenuated expression of CALD1 in SK-OV-3 cells leads to fewer F-actin stress fibers, reducing the association between the cytoskeleton and Vinculin. This results in reduced cellular focal adhesions and increased invasiveness of SK-OV-3 cells, promoting OV cell metastasis. These findings suggest that CALD1 may have important clinical implications in the diagnosis and treatment of OV.
Collapse
Affiliation(s)
- Yongchao Li
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar, China
| | - Zhao Yang
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Menglong Xu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar, China
| | - Haocheng Guan
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar, China
| | - Zhenhui Wu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar, China
| | - Shuwei Li
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar, China
| |
Collapse
|
2
|
Blagg BS, Catalfano KC. The role of Aha1 in cancer and neurodegeneration. Front Mol Neurosci 2024; 17:1509280. [PMID: 39776493 PMCID: PMC11703849 DOI: 10.3389/fnmol.2024.1509280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
The 90 kDa Heat shock protein (Hsp90) is a family of ubiquitously expressed molecular chaperones responsible for the stabilization and maturation of >400 client proteins. Hsp90 exhibits dramatic conformational changes to accomplish this, which are regulated by partner proteins termed co-chaperones. One of these co-chaperones is called the activator or Hsp90 ATPase activity homolog 1 (Aha1) and is the most potent accelerator of Hsp90 ATPase activity. In conditions where Aha1 levels are dysregulated including cystic fibrosis, cancer and neurodegeneration, Hsp90 mediated client maturation is disrupted. Accumulating evidence has demonstrated that many disease states exhibit large hetero-protein complexes with Hsp90 as the center. Many of these include Aha1, where increased Aha1 levels drive disease states forward. One strategy to block these effects is to design small molecule disruptors of the Hsp90/Aha1 complex. Studies have demonstrated that current Hsp90/Aha1 small molecule disruptors are effective in both models for cancer and neurodegeration.
Collapse
Affiliation(s)
- Brian S.J. Blagg
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
| | | |
Collapse
|
3
|
Romashin D, Rusanov A, Arzumanian V, Varshaver A, Poverennaya E, Vakhrushev I, Netrusov A, Luzgina N. Exploring the Functions of Mutant p53 through TP53 Knockout in HaCaT Keratinocytes. Curr Issues Mol Biol 2024; 46:1451-1466. [PMID: 38392212 PMCID: PMC10887868 DOI: 10.3390/cimb46020094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Approximately 50% of tumors carry mutations in TP53; thus, evaluation of the features of mutant p53 is crucial to understanding the mechanisms underlying cell transformation and tumor progression. HaCaT keratinocytes represent a valuable model for research in this area since they are considered normal, although they bear two gain-of-function mutations in TP53. In the present study, transcriptomic and proteomic profiling were employed to examine the functions of mutant p53 and to investigate the impact of its complete abolishment. Our findings indicate that CRISPR-mediated TP53 knockout results in significant changes at the transcriptomic and proteomic levels. The knockout of TP53 significantly increased the migration rate and altered the expression of genes associated with invasion, migration, and EMT but suppressed the epidermal differentiation program. These outcomes suggest that, despite being dysfunctional, p53 may still possess oncosuppressive functions. However, despite being considered normal keratinocytes, HaCaT cells exhibit oncogenic properties.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alexander Netrusov
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- Faculty of Biology and Biotechnology, HSE University, Moscow 101000, Russia
| | | |
Collapse
|
4
|
Fang M, Liu X, Xu W, Wang X, Xu L, Zhao TJ, Li P, Yang H. Paxillin family proteins Hic-5 and LPXN promote lipid storage by regulating the ubiquitination degradation of CIDEC. J Biol Chem 2024; 300:105610. [PMID: 38159847 PMCID: PMC10850781 DOI: 10.1016/j.jbc.2023.105610] [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: 10/10/2023] [Revised: 12/16/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Abstract
Many metabolic diseases are caused by disorders of lipid homeostasis. CIDEC, a lipid droplet (LD)-associated protein, plays a critical role in controlling LD fusion and lipid storage. However, regulators of CIDEC remain largely unknown. Here, we established a homogeneous time-resolved fluorescence (HTRF)-based high-throughput screening method and identified LPXN as a positive regulatory candidate for CIDEC. LPXN and Hic-5, the members of the Paxillin family, are focal adhesion adaptor proteins that contribute to the recruitment of specific kinases and phosphatases, cofactors, and structural proteins, participating in the transduction of extracellular signals into intracellular responses. Our data showed that Hic-5 and LPXN significantly increased the protein level of CIDEC and enhanced CIDEC stability not through triacylglycerol synthesis and FAK signaling pathways. Hic-5 and LPXN reduced the ubiquitination of CIDEC and inhibited its proteasome degradation pathway. Furthermore, Hic-5 and LPXN enlarged LDs and promoted lipid storage in adipocytes. Therefore, we identified Hic-5 and LPXN as novel regulators of CIDEC. Our current findings also suggest intervention with Hic-5 and LPXN might ameliorate ectopic fat storage by enhancing the lipid storage capacity of white adipose tissues.
Collapse
Affiliation(s)
- Mingyu Fang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Xu Liu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Wenbo Xu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Xing Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Lin Xu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Tong-Jin Zhao
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China; Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Shanghai Qi Zhi Institute, Shanghai, China
| | - Peng Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China; Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Shanghai Qi Zhi Institute, Shanghai, China
| | - Hui Yang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Virtanen V, Paunu K, Kukkula A, Niva S, Junila Y, Toriseva M, Jokilehto T, Mäkelä S, Huhtaniemi R, Poutanen M, Paatero I, Sundvall M. Glucocorticoid receptor-induced non-muscle caldesmon regulates metastasis in castration-resistant prostate cancer. Oncogenesis 2023; 12:42. [PMID: 37573448 PMCID: PMC10423232 DOI: 10.1038/s41389-023-00485-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/14/2023] Open
Abstract
Lethal prostate cancer (PCa) is characterized by the presence of metastases and development of resistance to therapies. Metastases form in a multi-step process enabled by dynamic cytoskeleton remodeling. An actin cytoskeleton regulating gene, CALD1, encodes a protein caldesmon (CaD). Its isoform, low-molecular-weight CaD (l-CaD), operates in non-muscle cells, supporting the function of filaments involved in force production and mechanosensing. Several factors, including glucocorticoid receptor (GR), have been identified as regulators of l-CaD in different cell types, but the regulation of l-CaD in PCa has not been defined. PCa develops resistance in response to therapeutic inhibition of androgen signaling by multiple strategies. Known strategies include androgen receptor (AR) alterations, modified steroid synthesis, and bypassing AR signaling, for example, by GR upregulation. Here, we report that in vitro downregulation of l-CaD promotes epithelial phenotype and reduces spheroid growth in 3D, which is reflected in vivo in reduced formation of metastases in zebrafish PCa xenografts. In accordance, CALD1 mRNA expression correlates with epithelial-to-mesenchymal transition (EMT) transcripts in PCa patients. We also show that CALD1 is highly co-expressed with GR in multiple PCa data sets, and GR activation upregulates l-CaD in vitro. Moreover, GR upregulation associates with increased l-CaD expression after the development of resistance to antiandrogen therapy in PCa xenograft mouse models. In summary, GR-regulated l-CaD plays a role in forming PCa metastases, being clinically relevant when antiandrogen resistance is attained by the means of bypassing AR signaling by GR upregulation.
Collapse
Affiliation(s)
- Verneri Virtanen
- Cancer Research Unit, Institute of Biomedicine, and FICAN West Cancer Center Laboratory, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Kreetta Paunu
- Cancer Research Unit, Institute of Biomedicine, and FICAN West Cancer Center Laboratory, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Antti Kukkula
- Cancer Research Unit, Institute of Biomedicine, and FICAN West Cancer Center Laboratory, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Saana Niva
- Cancer Research Unit, Institute of Biomedicine, and FICAN West Cancer Center Laboratory, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Ylva Junila
- Cancer Research Unit, Institute of Biomedicine, and FICAN West Cancer Center Laboratory, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Mervi Toriseva
- Cancer Research Unit, Institute of Biomedicine, and FICAN West Cancer Center Laboratory, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Terhi Jokilehto
- Cancer Research Unit, Institute of Biomedicine, and FICAN West Cancer Center Laboratory, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Sari Mäkelä
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, and FICAN West Cancer Center, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Riikka Huhtaniemi
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, and FICAN West Cancer Center, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Matti Poutanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, and FICAN West Cancer Center, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Ilkka Paatero
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Maria Sundvall
- Cancer Research Unit, Institute of Biomedicine, and FICAN West Cancer Center Laboratory, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland.
- Department of Oncology, Turku University Hospital, PL52, 20521, Turku, Finland.
| |
Collapse
|
6
|
Zhang J, Ren Z, Zheng D, Song Z, Lin J, Luo Y, Zou X, Pan Y, Qi N, Li A, Liu X. AHSA1 Promotes Proliferation and EMT by Regulating ERK/CALD1 Axis in Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:4600. [PMID: 36230524 PMCID: PMC9562867 DOI: 10.3390/cancers14194600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/13/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the major causes of cancer-related death worldwide. AHSA1 as a chaperone of HSP90 promotes the maturation, stability, and degradation of related cancer-promoting proteins. However, the regulatory mechanism and biological function of AHSA1 in HCC are largely unknown. Actually, we found that AHSA1 was significantly upregulated in HCC tissues and cell lines and was notably correlated with the poor clinical characteristics and prognosis of HCC patients in this study. Furthermore, both in vitro and in vivo, gain- and loss-of-function studies demonstrated that AHSA1 promoted the proliferation, invasion, metastasis, and epithelial-mesenchymal transition (EMT) of HCC. Moreover, the mechanistic study indicated that AHSA1 recruited ERK1/2 and promoted the phosphorylation and inactivation of CALD1, while ERK1/2 phosphorylation inhibitor SCH772984 reversed the role of AHSA1 in the proliferation and EMT of HCC. Furthermore, we demonstrated that the knockdown of CALD1 reversed the inhibition of proliferation and EMT by knocking AHSA1 in HCC. We also illustrated a new molecular mechanism associated with AHSA1 in HCC that was independent of HSP90 and MEK1/2. In summary, AHSA1 may play an oncogenic role in HCC by regulating ERK/CALD1 axis and may serve as a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Jiakang Zhang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
| | - Zhixuan Ren
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
| | - Dayong Zheng
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
| | - Zhenghui Song
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Junhao Lin
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
| | - Yue Luo
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
| | - Xiaopei Zou
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
| | - Yingying Pan
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
| | - Na Qi
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
- Department of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Aimin Li
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
| | - Xinhui Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
- Cancer Center, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
7
|
Alnuaimi AR, Nair VA, Malhab LJB, Abu-Gharbieh E, Ranade AV, Pintus G, Hamad M, Busch H, Kirfel J, Hamoudi R, Abdel-Rahman WM. Emerging role of caldesmon in cancer: A potential biomarker for colorectal cancer and other cancers. World J Gastrointest Oncol 2022; 14:1637-1653. [PMID: 36187394 PMCID: PMC9516648 DOI: 10.4251/wjgo.v14.i9.1637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/05/2022] [Accepted: 07/26/2022] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is a devastating disease, mainly because of metastasis. As a result, there is a need to better understand the molecular basis of invasion and metastasis and to identify new biomarkers and therapeutic targets to aid in managing these tumors. The actin cytoskeleton and actin-binding proteins are known to play an important role in the process of cancer metastasis because they control and execute essential steps in cell motility and contractility as well as cell division. Caldesmon (CaD) is an actin-binding protein encoded by the CALD1 gene as multiple transcripts that mainly encode two protein isoforms: High-molecular-weight CaD, expressed in smooth muscle, and low-molecular weight CaD (l-CaD), expressed in nonsmooth muscle cells. According to our comprehensive review of the literature, CaD, particularly l-CaD, plays a key role in the development, metastasis, and resistance to chemoradiotherapy in colorectal, breast, and urinary bladder cancers and gliomas, among other malignancies. CaD is involved in many aspects of the carcinogenic hallmarks, including epithelial mesenchymal transition via transforming growth factor-beta signaling, angiogenesis, resistance to hormonal therapy, and immune evasion. Recent data show that CaD is expressed in tumor cells as well as in stromal cells, such as cancer-associated fibroblasts, where it modulates the tumor microenvironment to favor the tumor. Interestingly, CaD undergoes selective tumor-specific splicing, and the resulting isoforms are generally not expressed in normal tissues, making these transcripts ideal targets for drug design. In this review, we will analyze these features of CaD with a focus on CRC and show how the currently available data qualify CaD as a potential candidate for targeted therapy in addition to its role in the diagnosis and prognosis of cancer.
Collapse
Affiliation(s)
- Alya R Alnuaimi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Vidhya A Nair
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Lara J Bou Malhab
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Eman Abu-Gharbieh
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Anu Vinod Ranade
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Basic Medical Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Gianfranco Pintus
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
| | - Mohamad Hamad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hauke Busch
- University Cancer Center Schleswig-Holstein and Luebeck Institute for Experimental Dermatology, University of Luebeck, Luebeck 23560, Germany
| | - Jutta Kirfel
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck 23560, Germany
| | - Rifat Hamoudi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London WC1E 6BT, United Kingdom
| | - Wael M Abdel-Rahman
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
8
|
Protein tyrosine kinase 2b inhibition reverts niche-associated resistance to tyrosine kinase inhibitors in AML. Leukemia 2022; 36:2418-2429. [PMID: 36056084 PMCID: PMC9522596 DOI: 10.1038/s41375-022-01687-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/08/2022]
Abstract
FLT3 tyrosine kinase inhibitor (TKI) therapy evolved into a standard therapy in FLT3-mutated AML. TKI resistance, however, develops frequently with poor outcomes. We analyzed acquired TKI resistance in AML cell lines by multilayered proteome analyses. Leupaxin (LPXN), a regulator of cell migration and adhesion, was induced during early resistance development, alongside the tyrosine kinase PTK2B which phosphorylated LPXN. Resistant cells differed in cell adhesion and migration, indicating altered niche interactions. PTK2B and LPXN were highly expressed in leukemic stem cells in FLT3-ITD patients. PTK2B/FAK inhibition abrogated resistance-associated phenotypes, such as enhanced cell migration. Altered pathways in resistant cells, assessed by nascent proteomics, were largely reverted upon PTK2B/FAK inhibition. PTK2B/FAK inhibitors PF-431396 and defactinib synergized with different TKIs or daunorubicin in FLT3-mutated AML. Midostaurin-resistant and AML cells co-cultured with mesenchymal stroma cells responded particularly well to PTK2B/FAK inhibitor addition. Xenograft mouse models showed significant longer time to leukemia symptom-related endpoint upon gilteritinib/defactinib combination treatment in comparison to treatment with either drug alone. Our data suggest that the leupaxin-PTK2B axis plays an important role in acquired TKI resistance in AML. PTK2B/FAK inhibitors act synergistically with currently used therapeutics and may overcome emerging TKI resistance in FLT3-mutated AML at an early timepoint.
Collapse
|
9
|
Khan A, Li W, Ambreen A, Wei DQ, Wang Y, Mao Y. A protein coupling and molecular simulation analysis of the clinical mutants of androgen receptor revealed a higher binding for Leupaxin, to increase the prostate cancer invasion and motility. Comput Biol Med 2022; 146:105537. [DOI: 10.1016/j.compbiomed.2022.105537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/19/2022]
|
10
|
Schmidt T. S-Adenosylmethionine affects ERK1/2 and STAT3 pathway in androgen-independent prostate cancer cells. Mol Biol Rep 2022; 49:4805-4817. [PMID: 35303200 PMCID: PMC9262802 DOI: 10.1007/s11033-022-07331-2] [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: 10/06/2021] [Accepted: 03/02/2022] [Indexed: 12/02/2022]
Abstract
Background The most critical point in the treatment of prostate cancer is the progression towards a hormone-refractory tumour, making research on alternative therapies necessary. This study focused on the methyl donor S-adenosylmethionine (SAM), which is known to act as an antitumourigenic in several cancer cell lines. Though a genome-wide downregulation of proto-oncogenes in prostate cancer cell lines treated with SAM is obvious, the anticancer effects remain elusive. Thus, in this study, the impact of SAM treatment on the cell cycle, apoptosis and cancer-related pathways was investigated. Methods and results After performing SAM treatment on prostate cancer cell lines (PC-3 and DU145), a cell-cycle arrest during the S-phase, a downregulation of cyclin A protein levels and an upregulation of p21 cell cycle inhibitor were observed. The proapoptotic Bax/Bcl-2 ratio and the caspase-3 activity were elevated; additionally, the apoptosis rate of SAM treated cells increased significantly in a time-dependent manner. Moreover, immunoblots displayed a downregulation of Erk1/2 and STAT3 phosphorylation accompanied by a reduced expression of the STAT3 protein. Conclusion SAM caused changes in cancer-related pathways, probably leading to the effects on the cell cycle and apoptosis rate. These results provide deeper insights into the anticancer effects of SAM on prostate cancer cells.
Collapse
Affiliation(s)
- Thomas Schmidt
- Institute of Anatomy and Clinical Morphology, University of Witten/Herdecke, 58448, Witten, Germany. .,Department of Anatomy and Developmental Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Strasse 7-11, Mannheim, Germany.
| |
Collapse
|
11
|
Yao YB, Xiao CF, Lu JG, Wang C. Caldesmon: Biochemical and Clinical Implications in Cancer. Front Cell Dev Biol 2021; 9:634759. [PMID: 33681215 PMCID: PMC7930484 DOI: 10.3389/fcell.2021.634759] [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: 11/28/2020] [Accepted: 01/18/2021] [Indexed: 12/19/2022] Open
Abstract
Caldesmon, an actin-binding protein, can inhibit myosin binding to actin and regulate smooth muscle contraction and relaxation. However, caldesmon has recently attracted attention due to its importance in cancer. The upregulation of caldesmon in several solid cancer tissues has been reported. Caldesmon, as well as its two isoforms, is considered as a biomarker for cancer and a potent suppressor of cancer cell invasion by regulating podosome/invadopodium formation. Therefore, caldesmon may be a promising therapeutic target for diseases such as cancer. Here, we review new studies on the gene transcription, isoform structure, expression, and phosphorylation regulation of caldesmon and discuss its clinical implications in cancer.
Collapse
Affiliation(s)
- Yi-Bo Yao
- Department of Anorectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang-Fang Xiao
- Department of Anorectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jin-Gen Lu
- Longhua Hospital, Institute of Chinese Traditional Surgery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Wang
- Department of Anorectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Yin X, Zhang X, Liu Z, Sun G, Zhu X, Zhang H, Zhu S, Zhao J, Chen J, Shen P, Wang J, Chen N, Zhou Q, Zeng H. Assessment for prognostic value of differentially expressed genes in immune microenvironment of clear cell renal cell carcinoma. Am J Transl Res 2020; 12:5416-5432. [PMID: 33042428 PMCID: PMC7540144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 08/01/2020] [Indexed: 06/11/2023]
Abstract
Tumor-infiltrating immune cells have been recognized to be associated with prognosis and response to immunotherapy; however, genes related to immune microenvironment of clear cell renal cell carcinoma (ccRCC) remains unclear. To better understand the effects of genes involved in immune and stromal cells on prognosis, we used Cancer Genome Atlas Kidney Renal Clear Cell Carcinoma (TCGA-KIRC), DAVID database and ESTMATE algorithm, and divided the patients into low and high groups according to immune (median: 1038.45) and stromal scores (median: 667.945), respectively. We found the immune scores were significantly correlated with clinicopathological parameters and overall survival (OS). Based on immune scores, 890 DEGs were significantly associated with OS among the 1433 up-regulated genes. Based on top 10 DEGs (IL10RA, FCER1G, SASH3, TIGIT, RHOH, IL12RB1, AIF1, LPXN, LAPTM5 and SP140), cases with number of up-regulated genes ≥ 5 were associated poor OS (P = 0.002). In addition, the mean differences of percentages of CD8 T cells (11.32%), CD4 memory resting T cells (-4.52%) and mast resting cells (-3.55%) between low and high immune scores were the most significant. Thus, combination of these genes might use to predict the efficacy of immunotherapy. Further analyses of these genes were warrant to explore their potential association with the prognosis of ccRCC.
Collapse
Affiliation(s)
- Xiaoxue Yin
- Department of Pathology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Pathology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Xingming Zhang
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Zhenhua Liu
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Guangxi Sun
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Xudong Zhu
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Haoran Zhang
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Sha Zhu
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Jinge Zhao
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Junru Chen
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Pengfei Shen
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Jia Wang
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Ni Chen
- Department of Pathology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Pathology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Qiao Zhou
- Department of Pathology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Pathology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| | - Hao Zeng
- Department of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
- Institute of Urology, West China Hospital, Sichuan UniversityChengdu 610041, P. R. China
| |
Collapse
|
13
|
Bonaud A, Clare S, Bisio V, Sowerby JM, Yao S, Ostergaard H, Balabanian K, Smith KGC, Espéli M. Leupaxin Expression Is Dispensable for B Cell Immune Responses. Front Immunol 2020; 11:466. [PMID: 32269569 PMCID: PMC7109257 DOI: 10.3389/fimmu.2020.00466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 02/28/2020] [Indexed: 11/22/2022] Open
Abstract
The generation of a potent humoral immune response by B cells relies on the integration of signals induced by the B cell receptor, toll-like receptors and both negative and positive co-receptors. Several reports also suggest that integrin signaling plays an important role in this process. How integrin signaling is regulated in B cells is however still partially understood. Integrin activity and function are controlled by several mechanisms including regulation by molecular adaptors of the paxillin family. In B cells, Leupaxin (Lpxn) is the most expressed member of the family and in vitro studies suggest that it could dampen BCR signaling. Here, we report that Lpxn expression is increased in germinal center B cells compared to naïve B cells. Moreover, Lpxn deficiency leads to decreased B cell differentiation into plasma cells in vitro. However, Lpxn seems dispensable for the generation of a potent B cell immune response in vivo. Altogether our results suggest that Lpxn is dispensable for T-dependent and T-independent B cell immune responses.
Collapse
Affiliation(s)
- Amélie Bonaud
- Inflammation Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Medicale (INSERM), Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Clamart, France
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France
| | - Simon Clare
- Wellcome Trust Genome, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Valeria Bisio
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France
| | - John M. Sowerby
- The Department of Medicine, Cambridge Biomedical, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Jeffrey Cheah Biomedical Centre Cambridge Biomedical, Cambridge Institute of Therapeutic Immunology & Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - Shugang Yao
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Hanne Ostergaard
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Karl Balabanian
- Inflammation Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Medicale (INSERM), Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Clamart, France
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France
| | - Kenneth G. C. Smith
- The Department of Medicine, Cambridge Biomedical, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Jeffrey Cheah Biomedical Centre Cambridge Biomedical, Cambridge Institute of Therapeutic Immunology & Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - Marion Espéli
- Inflammation Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Medicale (INSERM), Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Clamart, France
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France
| |
Collapse
|
14
|
Faria M, Shepherd P, Pan Y, Chatterjee SS, Navone N, Gustafsson JÅ, Strom A. The estrogen receptor variants β2 and β5 induce stem cell characteristics and chemotherapy resistance in prostate cancer through activation of hypoxic signaling. Oncotarget 2018; 9:36273-36288. [PMID: 30555629 PMCID: PMC6284737 DOI: 10.18632/oncotarget.26345] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/31/2018] [Indexed: 12/28/2022] Open
Abstract
Chemotherapy resistant prostate cancer is a major clinical problem. When the prostate cancer has become androgen deprivation resistant, one of the few treatment regimens left is chemotherapy. There is a strong connection between a cancer's stem cell like characteristics and drug resistance. By performing RNA-seq we observed several factors associated with stem cells being strongly up-regulated by the estrogen receptor β variants, β2 and β5. In addition, most of these factors were also up-regulated by hypoxia. One mechanism of chemotherapy resistance was expression of the hypoxia-regulated, drug transporter genes, where especially ABCG2 and MDR1 were shown to be expressed in recurrent prostate cancer and to cause chemotherapy resistance by efficiently transporting drugs like docetaxel out of the cells. Another mechanism was expression of the hypoxia-regulated Notch3 gene, which causes chemotherapy resistance in urothelial carcinoma, although the mechanism is unknown. It is well known that hypoxic signaling is involved in increasing chemotherapy resistance. Regulation of the hypoxic factors, HIF-1α and HIF-2α is very complex and extends far beyond hypoxia itself. We have recently shown that two of the estrogen receptor β variants, estrogen receptor β2 and β5, bind to and stabilize both HIF-1α and HIF-2α proteins leading to expression of HIF target genes. This study suggests that increased expression of the estrogen receptor β variants, β2 and β5, could be involved in development of a cancer's stem cell characteristics and chemotherapy resistance, indicating that targeting these factors could prevent or reverse chemotherapy resistance and cancer stem cell expansion.
Collapse
Affiliation(s)
- Michelle Faria
- University of Houston, Department of Biology and Biochemistry, Center for Nuclear, Receptors and Cell Signaling, Science and Engineering Research Center, Houston, Texas, USA
| | - Peter Shepherd
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yinghong Pan
- University of Houston, Department of Biology and Biochemistry, Center for Nuclear, Receptors and Cell Signaling, Science and Engineering Research Center, Houston, Texas, USA
| | - Sujash S Chatterjee
- University of Houston, Department of Biology and Biochemistry, Center for Nuclear, Receptors and Cell Signaling, Science and Engineering Research Center, Houston, Texas, USA
| | - Nora Navone
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jan-Åke Gustafsson
- University of Houston, Department of Biology and Biochemistry, Center for Nuclear, Receptors and Cell Signaling, Science and Engineering Research Center, Houston, Texas, USA.,Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden
| | - Anders Strom
- University of Houston, Department of Biology and Biochemistry, Center for Nuclear, Receptors and Cell Signaling, Science and Engineering Research Center, Houston, Texas, USA
| |
Collapse
|
15
|
Cheema AK, Byrum SD, Sharma NK, Altadill T, Kumar VP, Biswas S, Balgley BM, Hauer-Jensen M, Tackett AJ, Ghosh SP. Proteomic Changes in Mouse Spleen after Radiation-Induced Injury and its Modulation by Gamma-Tocotrienol. Radiat Res 2018; 190:449-463. [PMID: 30070965 PMCID: PMC6297072 DOI: 10.1667/rr15008.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gamma-tocotrienol (GT3), a naturally occurring vitamin E isomer, a promising radioprotector, has been shown to protect mice against radiation-induced hematopoietic and gastrointestinal injuries. We analyzed changes in protein expression profiles of spleen tissue after GT3 treatment in mice exposed to gamma radiation to gain insights into the molecular mechanism of radioprotective efficacy. Male CD2F1 mice, 12-to-14 weeks old, were treated with either vehicle or GT3 at 24 h prior to 7 Gy total-body irradiation. Nonirradiated vehicle, nonirradiated GT3 and age-matched naïve animals were used as controls. Blood and tissues were harvested on days 0, 1, 2, 4, 7, 10 and 14 postirradiation. High-resolution mass-spectrometry-based radioproteomics was used to identify differentially expressed proteins in spleen tissue with or without drug treatment. Subsequent bioinformatic analyses helped delineate molecular markers of biological pathways and networks regulating the cellular radiation responses in spleen. Our results show a robust alteration in spleen proteomic profiles including upregulation of the Wnt signaling pathway and actin-cytoskeleton linked proteins in mediating the radiation injury response in spleen. Furthermore, we show that 24 h pretreatment with GT3 attenuates radiation-induced hematopoietic injury in the spleen by modulating various cell signaling proteins. Taken together, our results show that the radioprotective effects of GT3 are mediated, via alleviation of radiation-induced alterations in biochemical pathways, with wide implications on overall hematopoietic injury.
Collapse
Affiliation(s)
- Amrita K. Cheema
- Departments of Oncology, Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Stephanie D. Byrum
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - Neel Kamal Sharma
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| | - Tatiana Altadill
- Departments of Oncology, Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC
- Institut d’Investigacio Biomedica de Bellvitge (IDIBELL), Gynecological Department, Vall Hebron University Hospital, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Vidya P. Kumar
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| | - Shukla Biswas
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| | | | - Martin Hauer-Jensen
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - Alan J. Tackett
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - Sanchita P. Ghosh
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| |
Collapse
|
16
|
Zhu GH, Dai HP, Shen Q, Zhang Q. Downregulation of LPXN expression by siRNA decreases the malignant proliferation and transmembrane invasion of SHI-1 cells. Oncol Lett 2018; 17:135-140. [PMID: 30655748 PMCID: PMC6313184 DOI: 10.3892/ol.2018.9605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 08/30/2018] [Indexed: 01/03/2023] Open
Abstract
The aim of the present study was to investigate the effects of decreasing leupaxin (LPXN) expression on the proliferation and invasion of human acute monocytic leukemia SHI-1 cells. The transfection efficiency of fluorescein amidite (FAM)-small interfering RNA (siRNA) was determined using flow cytometry, and the protein expression levels of LPXN, phosphorylated (p)-c-Jun N-terminal kinase (JNK), p-p38 mitogen-activated protein kinase (p38 MAPK) and p-extracellular-signal-regulated kinase (ERK) were detected by western blot analysis. Proliferation was determined using the cell counting kit-8 reagent and cellular transmembrane invasion ability was determined using a Transwell chamber system. The gelatinase levels of matrix metalloproteinase (MMP)-2 and MMP-9 in the cell culture supernatant were also analyzed by gelatin zymography. In SHI-1 cells, the optimal transfection conditions of siRNA were a cell density of 4×105 cells/ml and a ratio of siRNA/Lipofectamine® 2000 of 200 pmol/1 µl. The highest transfection efficiency of FAM-siRNA was 74.5%. In the present study, L2-siRNA was selected to effectively decrease the expression of LPXN. Following downregulation of LPXN expression by L2-siRNA, proliferation inhibition rates increased to 27.043±2.051 and cell transmembrane invasion rates decreased to 25.270±2.145 (P<0.05). The results of the western blot analysis and the gelatin zymography indicated that downregulation of LPXN expression increased the expression of p-p38 MAPK and p-JNK, and attenuated the secretion levels of MMP-2 and MMP-9. However, downregulation of LPXN expression had no effect on p-ERK expression in SHI-1 cells. The results of the present study indicated that downregulation of LPXN expression decreased the malignant proliferation and transmembrane invasion of SHI-1 cells by activating JNK and p38 MAPK, and inhibiting MMP-2 and MMP-9 secretion.
Collapse
Affiliation(s)
- Guo-Hua Zhu
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Hai-Ping Dai
- Leukemia Research Unit, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qun Shen
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China.,Department of Hematology, First Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210009, P.R. China
| | - Qi Zhang
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
17
|
朱 国, 戴 海, 段 元, 余 泽. [Small interfering RNA-mediated LPXN silencing suppresses proliferation and enhances drug sensitivity of human acute monocytic leukemia SHI-1 cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:807-811. [PMID: 33168498 PMCID: PMC6765540 DOI: 10.3969/j.issn.1673-4254.2018.07.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the effect of silencing LPXN expression by RNA interference on the proliferation and drug sensitivity of human acute monocytic leukemia SHI-1 cells in vitro. METHODS Small interfering RNA (siRNA) sequences targeting LPXN were designed and transiently transfected in SHI-1 cells via Lipofectamine 2000, and the most efficient siRNA sequence for LPXN silencing was identified using Western blotting. The protein expression levels of LPXN, p-JNK, p-P38 MAPK and p-ERK were in the cells transfected with the selected siRNA were detected using Western blotting, and the cell proliferation changes were assessed using CCK-8 reagent. RESULTS LPXN silencing by siRNA transfection resulted in significant proliferation suppression in SHI-1 cells with an inhibition rate of(27.04±2.05) % (P < 0.05). Western blotting showed that treatment of the siRNA-transfected SHI-1 cells with 0-25 μmol/L curcumin or with 0-2.0 μmol/L Ara-C further increased the cell inhibition rate and obviously enhanced the expressions of p-P38 MAPK and p-JNK without significantly affecting p-ERK expression. CONCLUSIONS Down-regulation of LPXN expression by siRNA transfection can suppress the proliferation and increase the drug sensitivity of SHI-1 cells probably by activating JNK and P38 MAPK.
Collapse
Affiliation(s)
- 国华 朱
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - 海萍 戴
- 苏州大学第一附属医院血液科,江苏 苏州 215006Department of Hematology, First Hospital Affiliated to Suzhou University, Suzhou 215006, China
| | - 元勋 段
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - 泽霖 余
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
18
|
Gao Y, Ge W. The histone methyltransferase DOT1L inhibits osteoclastogenesis and protects against osteoporosis. Cell Death Dis 2018; 9:33. [PMID: 29348610 PMCID: PMC5833786 DOI: 10.1038/s41419-017-0040-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/14/2017] [Accepted: 10/05/2017] [Indexed: 12/20/2022]
Abstract
Osteoclasts are absorptive cells that play a critical role in homeostatic bone remodeling and pathological bone resorption. Emerging evidence suggests an important role of epigenetic regulation in osteoclastogenesis. In this study, we investigated the role of DOT1L, which regulates gene expression epigenetically by histone H3K79 methylation (H3K79me), during osteoclast formation. Using RANKL-induced RAW264.7 macrophage cells as an osteoclast differentiation model, we found that DOT1L and H3K79me2 levels were upregulated during osteoclast differentiation. Small molecule inhibitor- (EPZ5676 or EPZ004777) or short hairpin RNA-mediated reduction in DOT1L expression promoted osteoclast differentiation and resorption. In addition, DOT1L inhibition increased osteoclast surface area and accelerated bone-mass reduction in a mouse ovariectomy (OVX) model of osteoporosis without alter osteoblast differentiation. DOT1L inhibition increase reactive oxygen species (ROS) generation and autophagy activity, and cell migration in pre-osteoclasts. Moreover, it strengthened expression of osteoclast fusion and resorption-related protein CD9 and MMP9 in osteoclasts derived from RAW264.7. Our findings support a new mechanism of DOT1L-regulated, H3K79me2-mediated, epigenetic regulation of osteoclast differentiation, implicating DOT1L as a new therapeutic target for osteoclast dysregulation-induced disease.
Collapse
Affiliation(s)
- Yanpan Gao
- State Key Laboratory of Medical Molecular Biology & Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, China.
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, China.
| |
Collapse
|
19
|
Chan CK, Pan Y, Nyberg K, Marra MA, Lim EL, Jones SJM, Maar D, Gibb EA, Gunaratne PH, Robertson AG, Rowat AC. Tumour-suppressor microRNAs regulate ovarian cancer cell physical properties and invasive behaviour. Open Biol 2016; 6:160275. [PMID: 27906134 PMCID: PMC5133448 DOI: 10.1098/rsob.160275] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022] Open
Abstract
The activities of pathways that regulate malignant transformation can be influenced by microRNAs (miRs). Recently, we showed that increased expression of five tumour-suppressor miRs, miR-508-3p, miR-508-5p, miR-509-3p, miR-509-5p and miR-130b-3p, correlate with improved clinical outcomes in human ovarian cancer patients, and that miR-509-3p attenuates invasion of ovarian cancer cell lines. Here, we investigate the mechanism underlying this reduced invasive potential by assessing the impact of these five miRs on the physical properties of cells. Human ovarian cancer cells (HEYA8, OVCAR8) that are transfected with miR mimics representing these five miRs exhibit decreased invasion through collagen matrices, increased cell size and reduced deformability as measured by microfiltration and microfluidic assays. To understand the molecular basis of altered invasion and deformability induced by these miRs, we use predicted and validated mRNA targets that encode structural and signalling proteins that regulate cell mechanical properties. Combined with analysis of gene transcripts by real-time PCR and image analysis of F-actin in single cells, our results suggest that these tumour-suppressor miRs may alter cell physical properties by regulating the actin cytoskeleton. Our findings provide biophysical insights into how tumour-suppressor miRs can regulate the invasive behaviour of ovarian cancer cells, and identify potential therapeutic targets that may be implicated in ovarian cancer progression.
Collapse
Affiliation(s)
- Clara K Chan
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Yinghong Pan
- Department of Biochemistry and Biology, University of Houston, Houston, TX, USA
| | - Kendra Nyberg
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Marco A Marra
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Emilia L Lim
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Steven J M Jones
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Dianna Maar
- Bio-Rad Laboratories, The Digital Biology Center, Pleasanton, CA, USA
| | - Ewan A Gibb
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Preethi H Gunaratne
- Department of Biochemistry and Biology, University of Houston, Houston, TX, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - A Gordon Robertson
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Amy C Rowat
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
20
|
Sand M, Bechara FG, Sand D, Gambichler T, Hahn SA, Bromba M, Stockfleth E, Hessam S. Circular RNA expression in basal cell carcinoma. Epigenomics 2016; 8:619-32. [DOI: 10.2217/epi-2015-0019] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: Circular RNAs (circRNAs), are nonprotein coding RNAs consisting of a circular loop with multiple miRNA, binding sites called miRNA response elements (MREs), functioning as miRNA sponges. This study was performed to identify differentially expressed circRNAs and their MREs in basal cell carcinoma (BCC). Materials & methods: Microarray circRNA expression profiles were acquired from BCC and control followed by qRT-PCR validation. Bioinformatical target prediction revealed multiple MREs. Sequence analysis was performed concerning MRE interaction potential with the BCC miRNome. Results: We identified 23 upregulated and 48 downregulated circRNAs with 354 miRNA response elements capable of sequestering miRNA target sequences of the BCC miRNome. Conclusion: The present study describes a variety of circRNAs that are potentially involved in the molecular pathogenesis of BCC.
Collapse
Affiliation(s)
- Michael Sand
- Dermatologic Surgery Unit, Department of Dermatology, Venereology & Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Plastic Surgery, St. Josef Hospital, Catholic Clinics of the Ruhr Peninsula, 45257 Essen, Germany
| | - Falk G Bechara
- Dermatologic Surgery Unit, Department of Dermatology, Venereology & Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Daniel Sand
- University of Michigan Kellogg Eye Center, Ann Arbor, MI 48105, USA
| | - Thilo Gambichler
- Dermatologic Surgery Unit, Department of Dermatology, Venereology & Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Stephan A Hahn
- Department of Internal Medicine, Knappschaftskrankenhaus University of Bochum, Zentrum für Klinische Forschung, Labor für Molekulare Gastroenterologische Onkologie, 44780 Bochum, Germany
| | - Michael Bromba
- Department of Plastic Surgery, St. Josef Hospital, Catholic Clinics of the Ruhr Peninsula, 45257 Essen, Germany
| | - Eggert Stockfleth
- Dermatologic Surgery Unit, Department of Dermatology, Venereology & Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Schapoor Hessam
- Dermatologic Surgery Unit, Department of Dermatology, Venereology & Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
| |
Collapse
|
21
|
Regulation of β-catenin transcription activity by leupaxin in hepatocellular carcinoma. Tumour Biol 2015; 37:2313-20. [PMID: 26361959 DOI: 10.1007/s13277-015-4060-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/04/2015] [Indexed: 01/06/2023] Open
Abstract
In human cancers, β-catenin is accumulated in the nucleus and activates mRNA transcription of many oncogenic genes, such as cyclin D1 and c-myc. However, the mechanism of β-catenin-mediated transcriptional activation remains largely unknown. In the present study, we identified leupaxin, an adaptor protein sharing homology with the focal adhesion protein, as a novel coactivator for β-catenin in human hepatocellular carcinoma (HCC). We show that leupaxin could interact with β-catenin and enhance its transcriptional activity through recruitment of coactivator complex, including steroid receptor coactivator 1 (SRC-1) and P300. As a result, leupaxin regulates HCC cell proliferation and cell-cycle progression in the presence of intact Wnt/β-catenin signaling. Furthermore, leupaxin is overexpressed in HCC tissues and correlated with mRNA levels of cyclin D1 and c-myc. Therefore, this is the first demonstration of a role for the leupaxin in the regulation of HCC progression, at least in part, by enhancing β-catenin transcription activity.
Collapse
|