1
|
Fan YJ, Pan FZ, Cui ZG, Zheng HC. The Antitumor and Sorafenib-resistant Reversal Effects of Ursolic Acid on Hepatocellular Carcinoma via Targeting ING5. Int J Biol Sci 2024; 20:4190-4208. [PMID: 39247819 PMCID: PMC11379078 DOI: 10.7150/ijbs.97720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/15/2024] [Indexed: 09/10/2024] Open
Abstract
Inhibitor of growth 5 (ING5) has been reported to be involved in the malignant progression of cancers. Ursolic acid (UA) has shown remarkable antitumor effects. However, its antitumor mechanisms regarding of ING5 in hepatocellular carcinoma (HCC) remain unclear. Herein, we found that UA significantly suppressed the proliferation, anti-apoptosis, migration and invasion of HCC cells. In addition, ING5 expression in HCC cells treated with UA was obviously downregulated in a concentration- and time-dependent manner. Additionally, the pro-oncogenic role of ING5 was confirmed in HCC cells. Further investigation revealed that UA exerted antitumor effects on HCC by inhibiting ING5-mediated activation of PI3K/Akt pathway. Notably, UA could also reverse sorafenib resistance of HCC cells by suppressing the ING5-ACC1/ACLY-lipid droplets (LDs) axis. UA abrogated ING5 transcription and downregulated its expression by reducing SRF and YY1 expression and the SRF-YY1 complex formation. Alb/JCPyV T antigen mice were used for in vivo experiments since T antigen upregulated ING5 expression by inhibiting the ubiquitin-mediated degradation and promoting the T antigen-SRF-YY1-ING5 complex-associated transcription. UA suppressed JCPyV T antigen-induced spontaneous HCC through inhibiting ING5-mediated PI3K/Akt signaling pathway. These findings suggest that UA has the dual antitumoral functions of inhibiting hepatocellular carcinogenesis and reversing sorafenib resistance of HCC cells through targeting ING5, which could serve as a potential therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Yin-Jie Fan
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang 110001, Liaoning Province, China
| | - Fu-Zhi Pan
- Department of Ultrasound Medicine, Liaoning Cancer Hospital, Shenyang 110001, Liaoning Province, China
| | - Zheng-Guo Cui
- Department of Environmental Health, University of Fukui School of Medical Sciences, Fukui 910-1193, Japan
| | - Hua-Chuan Zheng
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei Province, China
| |
Collapse
|
2
|
Chen S, Leng P, Guo J, Zhou H. FBXW7 in breast cancer: mechanism of action and therapeutic potential. J Exp Clin Cancer Res 2023; 42:226. [PMID: 37658431 PMCID: PMC10474666 DOI: 10.1186/s13046-023-02767-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/18/2023] [Indexed: 09/03/2023] Open
Abstract
Breast cancer is one of the frequent tumors that seriously endanger the physical and mental well-being in women. F-box and WD repeat domain-containing 7 (FBXW7) is a neoplastic repressor. Serving as a substrate recognition element for ubiquitin ligase, FBXW7 participates in the ubiquitin-proteasome system and is typically in charge of the ubiquitination and destruction of crucial oncogenic proteins, further performing a paramount role in cell differentiation, apoptosis and metabolic processes. Low levels of FBXW7 cause abnormal stability of pertinent substrates, mutations and/or deletions in the FBXW7 gene have been reported to correlate with breast cancer malignant progression and chemoresistance. Given the lack of an effective solution to breast cancer's clinical drug resistance dilemma, elucidating FBXW7's mechanism of action could provide a theoretical basis for targeted drug exploration. Therefore, in this review, we focused on FBXW7's role in a range of breast cancer malignant behaviors and summarized the pertinent cellular targets, signaling pathways, as well as the mechanisms regulating FBXW7 expression. We also proposed novel perspectives for the exploitation of alternative therapies and specific tumor markers for breast cancer by therapeutic strategies aiming at FBXW7.
Collapse
Affiliation(s)
- Siyu Chen
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosisand, Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology , Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Leng
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosisand, Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology , Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlin Guo
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosisand, Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology , Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Hao Zhou
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosisand, Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology , Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
3
|
Enríquez-Flores S, De la Mora-De la Mora I, García-Torres I, Flores-López LA, Martínez-Pérez Y, López-Velázquez G. Human Triosephosphate Isomerase Is a Potential Target in Cancer Due to Commonly Occurring Post-Translational Modifications. Molecules 2023; 28:6163. [PMID: 37630415 PMCID: PMC10459230 DOI: 10.3390/molecules28166163] [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: 06/14/2023] [Revised: 08/04/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer involves a series of diseases where cellular growth is not controlled. Cancer is a leading cause of death worldwide, and the burden of cancer incidence and mortality is rapidly growing, mainly in developing countries. Many drugs are currently used, from chemotherapeutic agents to immunotherapy, among others, along with organ transplantation. Treatments can cause severe side effects, including remission and progression of the disease with serious consequences. Increased glycolytic activity is characteristic of cancer cells. Triosephosphate isomerase is essential for net ATP production in the glycolytic pathway. Notably, some post-translational events have been described that occur in human triosephosphate isomerase in which functional and structural alterations are provoked. This is considered a window of opportunity, given the differences that may exist between cancer cells and their counterpart in normal cells concerning the glycolytic enzymes. Here, we provide elements that bring out the potential of triosephosphate isomerase, under post-translational modifications, to be considered an efficacious target for treating cancer.
Collapse
Affiliation(s)
- Sergio Enríquez-Flores
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (I.D.l.M.-D.l.M.); (I.G.-T.)
| | - Ignacio De la Mora-De la Mora
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (I.D.l.M.-D.l.M.); (I.G.-T.)
| | - Itzhel García-Torres
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (I.D.l.M.-D.l.M.); (I.G.-T.)
| | - Luis A. Flores-López
- Laboratorio de Biomoléculas y Salud Infantil, CONAHCYT-Instituto Nacional de Pediatría, Mexico City 04530, Mexico;
| | - Yoalli Martínez-Pérez
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Mexico City 14380, Mexico;
| | - Gabriel López-Velázquez
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (I.D.l.M.-D.l.M.); (I.G.-T.)
| |
Collapse
|
4
|
Murillo Carrasco AG, Giovanini G, Ramos AF, Chammas R, Bustos SO. Insights from a Computational-Based Approach for Analyzing Autophagy Genes across Human Cancers. Genes (Basel) 2023; 14:1550. [PMID: 37628602 PMCID: PMC10454514 DOI: 10.3390/genes14081550] [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: 06/30/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
In the last decade, there has been a boost in autophagy reports due to its role in cancer progression and its association with tumor resistance to treatment. Despite this, many questions remain to be elucidated and explored among the different tumors. Here, we used omics-based cancer datasets to identify autophagy genes as prognostic markers in cancer. We then combined these findings with independent studies to further characterize the clinical significance of these genes in cancer. Our observations highlight the importance of innovative approaches to analyze tumor heterogeneity, potentially affecting the expression of autophagy-related genes with either pro-tumoral or anti-tumoral functions. In silico analysis allowed for identifying three genes (TBC1D12, KERA, and TUBA3D) not previously described as associated with autophagy pathways in cancer. While autophagy-related genes were rarely mutated across human cancers, the expression profiles of these genes allowed the clustering of different cancers into three independent groups. We have also analyzed datasets highlighting the effects of drugs or regulatory RNAs on autophagy. Altogether, these data provide a comprehensive list of targets to further the understanding of autophagy mechanisms in cancer and investigate possible therapeutic targets.
Collapse
Affiliation(s)
- Alexis Germán Murillo Carrasco
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (S.O.B.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Guilherme Giovanini
- Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, Av. Arlindo Béttio, 1000, São Paulo 03828-000, Brazil; (G.G.); (A.F.R.)
| | - Alexandre Ferreira Ramos
- Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, Av. Arlindo Béttio, 1000, São Paulo 03828-000, Brazil; (G.G.); (A.F.R.)
| | - Roger Chammas
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (S.O.B.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Silvina Odete Bustos
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (S.O.B.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| |
Collapse
|
5
|
Zheng HC, Xue H, Jiang HM. The roles of ING5 in cancer: A tumor suppressor. Front Cell Dev Biol 2022; 10:1012179. [PMID: 36425530 PMCID: PMC9679416 DOI: 10.3389/fcell.2022.1012179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/05/2022] [Indexed: 11/09/2022] Open
Abstract
As a Class II tumor suppressor, ING5 contains nuclear localization signal, plant homeodomain, novel conserved region, and leucine zipper-like domains. ING5 proteins form homodimer into a coil-coil structure, and heterodimers with ING4, histone H3K4me3, histone acetyltransferase (HAT) complex, Tip60, Cyclin A1/CDK2, INCA1 and EBNA3C for the transcription of target genes. The acetylated proteins up-regulated by ING5 are preferentially located in nucleus and act as transcription cofactors, chromatin and DNA binding functions, while those down-regulated by ING5 mostly in cytoplasm and contribute to metabolism. ING5 promotes the autoacetylation of HAT p300, p53, histone H3 and H4 for the transcription of downstream genes (Bax, GADD45, p21, p27 and so forth). Transcriptionally, YY1 and SRF up-regulate ING5 mRNA expression by the interaction of YY1-SRF-p53-ING5 complex with ING5 promoter. Translationally, ING5 is targeted by miR-196, miR-196a, miR-196b-5p, miR-193a-3p, miR-27-3p, miR-200b/200a/429, miR-1307, miR-193, miR-222, miR-331-3p, miR-181b, miR-543 and miR-196-b. ING5 suppresses proliferation, migration, invasion and tumor growth of various cancer cells via the suppression of EGFR/PI3K/Akt, IL-6/STAT3, Akt/NF-κB/NF-κB/MMP-9 or IL-6/CXCL12 pathway. ING5-mediated chemoresistance is closely linked to anti-apoptosis, overexpression of chemoresistant genes, the activation of PI3K/Akt/NF-κB and Wnt/β-catenin signal pathways. Histologically, ING5 abrogation in gastric stem-like and pdx1-positive cells causes gastric dysplasia and cancer, and conditional ING5 knockout in pdx1-positive and gastric chief cells increases MNU-induced gastric carcinogenesis. Intestinal ING5 deletion increases AOM/DSS- induced colorectal carcinogenesis and decreases high-fat-diet weight. The overexpression and nucleocytoplasmic translocation of ING5 are seen during carcinogenesis, and ING5 expression was inversely associated with aggressive behaviors and poor prognosis in a variety of cancers. These findings indicated that ING5 might be used for a molecular marker for carcinogenesis and following progression, and as a target for gene therapy if its chemoresistant function might be ameliorated.
Collapse
Affiliation(s)
- Hua-chuan Zheng
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, China
- *Correspondence: Hua-chuan Zheng,
| | - Hang Xue
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Hua-mao Jiang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
6
|
Inhibitor of Growth Factors Regulate Cellular Senescence. Cancers (Basel) 2022; 14:cancers14133107. [PMID: 35804879 PMCID: PMC9264871 DOI: 10.3390/cancers14133107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Five members of the Inhibitor of Growth (ING) family share a highly conserved plant homeodomian with affinity to the specific histone modification H3K4me3. Since some ING family members are preferentially associated with histone acetyltransferaseactivity while other members with histone deacetlyse activity, the ING family membres are epigenetic regulators. Interestingly, ING members can regulate the induction cellular senescence in both primray untransformed human cells as well as human cancer cells. We discuss here the up-to-date knowledge about their regulatory activity within the cellular senescent program. Abstract The Inhibitor of Growth (ING) proteins are a group of tumor suppressors with five conserved genes. A common motif of ING factors is the conserved plant homeodomain (PHD), with which they bind to chromatin as readers of the histone mark trimethylated histone H3 (H3K4me3). These genes often produce several protein products through alternative splicing events. Interestingly, ING1 and ING2 participate in the establishment of the repressive mSIN3a-HDAC complexes, whereas ING3, ING4, and ING5 are associated with the activating HAT protein complexes. In addition to the modulation of chromatin’s structure, they regulate cell cycle transition, cellular senescence, repair of DNA damage, apoptosis, and angiogenic pathways. They also have fundamental effects on regulating cellular senescence in cancer cells. In the current review, we explain their role in cellular senescence based on the evidence obtained from cell line and animal studies, particularly in the context of cancer.
Collapse
|
7
|
Zheng HC, Xue H, Wu X, Xu HL, Zhao EH, Cui ZG. Transcriptional Regulation of ING5 and its Suppressive Effects on Gastric Cancer. Front Oncol 2022; 12:918954. [PMID: 35747809 PMCID: PMC9209732 DOI: 10.3389/fonc.2022.918954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/12/2022] [Indexed: 01/23/2023] Open
Abstract
ING5 targets histone acetyltransferase or histone deacetylase complexes for local chromatin remodeling. Its transcriptional regulation and suppressive effects on gastric cancer remain elusive. Luciferase assay, EMSA, and ChIP were used to identify the cis-acting elements and trans-acting factors of the ING5 gene. We analyzed the effects of SAHA on the aggressive phenotypes of ING5 transfectants, and the effects of different ING5 mutants on aggressive phenotypes in SGC-7901 cells. Finally, we observed the effects of ING5 abrogation on gastric carcinogenesis. EMSA and ChIP showed that both SRF (−717 to −678 bp) and YY1 (−48 to 25bp) interacted with the promoter of ING5 and up-regulated ING5 expression in gastric cancer via SRF-YY1-ING5-p53 complex formation. ING5, SRF, and YY1 were overexpressed in gastric cancer, (P<0.05), and associated with worse prognosis of gastric cancer patients (P<0.05). ING5 had positive relationships with SRF and YY1 expression in gastric cancer (P<0.05). SAHA treatment caused early arrest at S phase in ING5 transfectants of SGC-7901 (P<0.05), and either 0.5 or 1.0 μM SAHA enhanced their migration and invasion (P<0.05). The wild-type and mutant ING5 transfectants showed lower viability and invasion than the control (P<0.05) with low CDC25, VEGF, and MMP-9 expression. Gastric spontaneous adenocarcinoma was observed in Atp4b-cre; ING5f/f, Pdx1-cre; ING5f/f, and K19-cre; ING5f/f mice. ING5 deletion increased the sensitivity of MNU-induced gastric carcinogenesis. ING5 mRNA might be a good marker of gastric carcinogenesis, and poor prognosis. ING5 expression was positively regulated by the interaction of SRF-YY1-ING5-p53 complex within the ING5 promoter from −50 bp upstream to the transcription start site. ING5 deletion might contribute to the tumorigenesis and histogenesis of gastric cancer.
Collapse
Affiliation(s)
- Hua-chuan Zheng
- Department of Oncology and Experimental Center, The Affiliated Hospital of Chengde Medical University, Chengde, China
- *Correspondence: Hua-chuan Zheng,
| | - Hang Xue
- Department of Oncology and Experimental Center, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Xin Wu
- Department of Pathology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Hai-lan Xu
- Department of Oncology and Experimental Center, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - En-hong Zhao
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Zheng-guo Cui
- Department of Environmental Health, University of Fukui School of Medical Science, Fukui, Japan
| |
Collapse
|
8
|
Jiang J, Wang HJ, Mou XZ, Zhang H, Chen Y, Hu ZM. Low Expression of KAT6B May Affect Prognosis in Hepatocellular Carcinoma. Technol Cancer Res Treat 2021; 20:15330338211033063. [PMID: 34464167 PMCID: PMC8411621 DOI: 10.1177/15330338211033063] [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] [Indexed: 12/24/2022] Open
Abstract
Aims: Lysine acetyltransferase 6B (KAT6B), is a histone acetyltransferase
implicated to have a role in tumor suppression. However, the relationship
between KAT6B and hepatocellular carcinoma (HCC) is unclear. The purpose of
this study was to detect the expression of KAT6B in HCC tissues and analyze
its connection with the clinicopathological features of HCC. Methods: First, we performed immunohistochemical staining on 250 HCC tissues and 222
non-tumor liver tissues to examine the expression of KAT6B.Then the relation
between KAT6B expression and clinicopathological parameters was analyzed by
chi-square test, and the overall survival analysis was conducted by
Kaplan-Meier survival method. In addition, based on the Oncomine expression
array online and the UALCAN database, we compared KAT6B expression
differences between normal liver tissues and HCC tissues more broadly. Results: Compared with normal tissues, KAT6B expression was significantly lower in HCC
tissues. Low KAT6B expression was found to be related to gender, AFP level,
and tumor size. According to the online database, KAT6B expression was found
to be decreased in HCC tissues and high in normal tissues. Conclusions: Lower expression of KAT6B is associated with poor prognosis of HCC, and KAT6B
may be a potential tumor suppressor in liver cancer.
Collapse
Affiliation(s)
- Junjie Jiang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China.,Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China
| | - Hui-Ju Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China
| | - Huanqing Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China.,Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China
| | - YiZhen Chen
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China.,Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China
| | - Zhi-Ming Hu
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China
| |
Collapse
|
9
|
Wu X, Chen C, Luo B, Yan D, Yan H, Chen F, Guan F, Wu H, Yuan J. Nuclear ING3 Expression Is Correlated With a Good Prognosis of Breast Cancer. Front Oncol 2021; 10:589009. [PMID: 33469513 PMCID: PMC7813678 DOI: 10.3389/fonc.2020.589009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/26/2020] [Indexed: 12/23/2022] Open
Abstract
The inhibitor of growth (ING) family was discovered as the type II tumor suppressors, which regulated the proliferation, apoptosis, differentiation, angiogenesis, metastasis, and invasion of tumor cells through multiple pathways. ING3, a new member of ING family, has been reported to be downregulated in several types of tumors. However, few studies on ING3 in breast cancer have been reported. In this study, we investigated the expression of ING3 and determined its prognostic value in breast cancer. The immunohistochemistry was performed to evaluate the expression of ING3 in tissue microarrays (TMA) including breast cancer tissues (n=211) and normal breast tissues (n=50). In normal breast tissues, ING3 protein was detected in both the cytoplasm and nucleus. In breast cancer tissues, ING3 protein was principally detected in the cytoplasm. Compared with normal breast tissues, the expression of ING3 in nucleus was remarkably reduced in breast cancer tissues. The downregulated ING3 in nucleus was significantly correlated with clinicopathological characteristics including histological grade, lymph node metastasis, and the status of ER and PR. In HER2 positive-type and triple-negative breast cancer (TNBC) patients, it had the lower rate of nuclear ING3 with high expression than that in luminal-type. Moreover, Kaplan-Meier curves demonstrated that the reduced expression of ING3 in nucleus was correlated with a poorer 5-DFS and 5-OS of breast cancer patients. Importantly, multivariate Cox regression analysis suggested that the reduced expression of ING3 in nucleus was an independent prognostic factor in breast cancer. Our study comprehensively described the expression of ING3 in breast cancer for the first time and proved that it was an independent prognostic predictor of breast cancer, as well as a new idea for study of breast cancer.
Collapse
Affiliation(s)
- Xiaoyan Wu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bin Luo
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dandan Yan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Honglin Yan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangfang Chen
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Guan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Wu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Zheng HC, Zhao S, Xue H, Zhao EH, Jiang HM, Hao CL. The Roles of Beclin 1 Expression in Gastric Cancer: A Marker for Carcinogenesis, Aggressive Behaviors and Favorable Prognosis, and a Target of Gene Therapy. Front Oncol 2020; 10:613679. [PMID: 33425768 PMCID: PMC7787063 DOI: 10.3389/fonc.2020.613679] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/17/2020] [Indexed: 12/21/2022] Open
Abstract
Beclin 1 is encoded by Becn1, and plays a role in tumorigenesis, neurodegeneration, apoptosis and autophagy. Here, the aggressive phenotypes and relevant proteins were examined after Beclin 1 expression was altered in gastric cancer cells. We also observed the effects of Beclin 1 on gastric carcinogenesis using Becn1 knockout mice. Finally, clinicopathological significances of Beclin 1 expression were analyzed using meta- and bioinformatics analyses. Becn1 overexpression was found to inhibit proliferation, glucose metabolism, migration and invasion of gastric cancer cells, whereas its knockdown caused the opposite effects. Beclin 1 suppressed the tumor growth by decreasing proliferation and increasing apoptosis. The heterozygous abrogation of Becn1 in gastric pit, parietal and chief cells could not cause any epithelial lesion. Beclin 1-mediated chemoresistance was closely linked to the autophagy, Bax underexpression, and the overexpression of Bcl-2, LRP1, MDR1, and ING5. Bioinformatics analysis showed higher Becn1 mRNA expression in intestinal- than diffuse-type carcinomas (P<0.05), and in male than female gastric cancer patients (P<0.05). Becn1 hyperexpression was positively associated with both overall and progression-free survival rates of the cancer patients (P<0.05). Meta-analysis showed that down-regulated Beclin 1 expression in gastric cancer was positively with lymph node metastasis, TNM staging, dedifferentiation and poor prognosis (P<0.05). Becn1-related signal pathways in gastric cancer included prostate, lung, renal, colorectal, endometrial and thyroid cancers, glioma, and leukemia, the metabolism of amino acid, lipid and sugar, and some signal pathways of insulin, MAPK, TRL, VEGF, JAK-STAT, chemokine, p53, lysosome, peroxidome and ubiquitin-mediated protein degradation (P<0.05). These suggested that Beclin 1 might be considered as a potential marker of gastric carcinogenesis, aggressiveness and prognostic prediction, and as a target of gene therapy in gastric cancer.
Collapse
Affiliation(s)
- Hua-Chuan Zheng
- Department of Oncology and Experimental Center, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Shuang Zhao
- Department of Oncology and Experimental Center, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Hang Xue
- Department of Oncology and Experimental Center, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - En-Hong Zhao
- Department of Surgery, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Hua-Mao Jiang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chang-Lai Hao
- Department of Hematology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| |
Collapse
|
11
|
Barlak N, Capik O, Sanli F, Kilic A, Aytatli A, Yazici A, Ortucu S, Ittmann M, Karatas OF. ING5 inhibits cancer aggressiveness by inhibiting Akt and activating p53 in prostate cancer. Cell Biol Int 2020; 44:242-252. [PMID: 31475765 DOI: 10.1002/cbin.11227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PCa) is one of the most common types of cancer in men. In several recent studies, chromosomal deletions in the q arm of chromosome 2, where ING5 resides within, have been identified in various cancer types including PCa. In this study, we investigate the role of ING5 as a tumor suppressor in PCa. We examined the expression level of ING5 in tissue samples and cell lines using quantitative real-time polymerase chain reaction and western blot analysis. We tested the in vitro tumor suppressor potential of ING5 in PC3 and LNCaP cells stably overexpressing it using cell viability, colony formation, migration, invasion, and apoptosis assays. We then investigated the effects of ING5 on the Akt and p53 signaling using western blot analysis. We show that ING5 is significantly downregulated in PCa tumor tissue samples and cell lines compared with the corresponding controls. In vitro assays demonstrate that ING5 effectively suppresses proliferative, clonogenic, migratory, and invasive potential and induce apoptosis in PCa cells. ING5 may potentially exert its anti-tumor potential by inhibiting AKT and inducing p53 signaling pathways. Our findings demonstrate that ING5 possesses tumor suppressor roles in vitro, pointing its importance during the prostatic carcinogenesis processes.
Collapse
Affiliation(s)
- Neslisah Barlak
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Ozel Capik
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Fatma Sanli
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Ahsen Kilic
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Abdulmelik Aytatli
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Aysenur Yazici
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Serkan Ortucu
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Michael Ittmann
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, 77030, USA.,Michael E. DeBakey VAMC, Houston, Texas, 77030, USA
| | - Omer Faruk Karatas
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| |
Collapse
|
12
|
Ormaza G, Rodríguez JA, Ibáñez de Opakua A, Merino N, Villate M, Gorroño I, Rábano M, Palmero I, Vilaseca M, Kypta R, Vivanco MDM, Rojas AL, Blanco FJ. The Tumor Suppressor ING5 Is a Dimeric, Bivalent Recognition Molecule of the Histone H3K4me3 Mark. J Mol Biol 2019; 431:2298-2319. [PMID: 31026448 DOI: 10.1016/j.jmb.2019.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/10/2019] [Accepted: 04/10/2019] [Indexed: 10/26/2022]
Abstract
The INhibitor of Growth (ING) family of tumor suppressors regulates the transcriptional state of chromatin by recruiting remodeling complexes to sites with histone H3 trimethylated at lysine 4 (H3K4me3). This modification is recognized by the plant homeodomain (PHD) present at the C-terminus of the five ING proteins. ING5 facilitates histone H3 acetylation by the HBO1 complex, and also H4 acetylation by the MOZ/MORF complex. We show that ING5 forms homodimers through its N-terminal domain, which folds independently into an elongated coiled-coil structure. The central region of ING5, which contains the nuclear localization sequence, is flexible and disordered, but it binds dsDNA with micromolar affinity. NMR analysis of the full-length protein reveals that the two PHD fingers of the dimer are chemically equivalent and independent of the rest of the molecule, and they bind H3K4me3 in the same way as the isolated PHD. We have observed that ING5 can form heterodimers with the highly homologous ING4, and that two of three primary tumor-associated mutants in the N-terminal domain strongly destabilize the coiled-coil structure. They also affect cell proliferation and cell cycle phase distribution, suggesting a driver role in cancer progression.
Collapse
Affiliation(s)
- Georgina Ormaza
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | | | | | - Nekane Merino
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Maider Villate
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Irantzu Gorroño
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Miriam Rábano
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Ignacio Palmero
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, 28029 Madrid, Spain
| | - Marta Vilaseca
- Institute for Research in Biomedicine, 08028 Barcelona, Spain
| | - Robert Kypta
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; Department of Surgery and Cancer, Imperial College London, London, W12 0NN, UK
| | | | - Adriana L Rojas
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Francisco J Blanco
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain.
| |
Collapse
|
13
|
Regulat-INGs in tumors and diseases: Focus on ncRNAs. Cancer Lett 2019; 447:66-74. [PMID: 30673590 DOI: 10.1016/j.canlet.2019.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/11/2022]
Abstract
ING family genes (Inhibitor of Growth) are tumor suppressor genes that play a vital role in cell homeostasis. It has been shown that their expression is lost or diminished in many cancers and other diseases. The main mechanisms by which they are regulated in oncogenesis have not yet been fully elucidated. The involvement of non-coding RNAs (ncRNAs) and in particular microRNAs (miRNAs) in post-transcriptional gene regulation is well established. miRNAs are short sequences (18-25 nucleotides) that can bind to the 3 'UTR sequence of the targeted messenger RNA (mRNA), leading to its degradation or translational repression. Interactions between the ING family and miRNAs have been described in some cancers but also in other diseases. The involvement of miRNAs in ING family regulation opens up new fields of investigation, particularly for targeted therapies. In this review, we will summarize the regulatory mechanisms at the RNA and protein level of the ING family and focus on the interactions with ncRNAs.
Collapse
|
14
|
Gong J, Zhou Y, Liu D, Huo J. F-box proteins involved in cancer-associated drug resistance. Oncol Lett 2018; 15:8891-8900. [PMID: 29805625 DOI: 10.3892/ol.2018.8500] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 01/24/2018] [Indexed: 12/11/2022] Open
Abstract
The ubiquitin proteasome system (UPS) regulated human biological processes through the appropriate and efficient proteolysis of cellular proteins. F-box proteins are the vital components of SKP1-CUL1-FBP (SCF)-type E3 ubiquitin ligases that determine substrate specificity. As F-box proteins have the ability to control the degradation of several crucial protein targets associated with drug resistance, the dysregulation of these proteins may lead to induction of chemoresistance in cancer cells. Chemotherapy is one of the most conventional therapeutic approaches of treatment of patients with cancer. However, its exclusive application in clinical settings is restricted due to the development of chemoresistance, which typically results treatment failure. Therefore, overcoming drug resistance is considered as one of the most critical issues that researchers and clinician associated with oncology face. The present review serves to provide a comprehensive overview of F-box proteins and their possible targets as well as their correlation with the chemoresistance and chemosensitization of cancer cells. The article also presents an integrated representation of the complex regulatory mechanisms responsible for chemoresistance, which may lay the foundation to explore sensible candidate drugs for therapeutic intervention.
Collapse
Affiliation(s)
- Jian Gong
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yuqian Zhou
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Deliang Liu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jirong Huo
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
15
|
Ye P, Ke X, Zang X, Sun H, Dong Z, Lin J, Wang L, Liu W, Miao G, Tan Y, Tong W, Xiao H, Gao L. Up-regulated MiR-27-3p promotes the G1-S phase transition by targeting inhibitor of growth family member 5 in osteosarcoma. Biomed Pharmacother 2018; 101:219-227. [PMID: 29494959 DOI: 10.1016/j.biopha.2018.02.066] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE MicroRNAs (miRNAs) play an essential role in regulating malignant progression of tumour cells by inhibiting translation or stability of messenger RNA. However, the expression pattern and regulatory mechanism of miR-27-3p in osteosarcoma remains unclear. METHODS We examined the expression of miR-27-3p in 5 osteosarcoma cell lines compared with that in 2 normal osteocyte cell lines. Osteosarcoma cells U-2OS and MG-63 were transduced to up-regulate or down-regulate the expression of miR-27-3p. The 3-(4, 5-Dimethyl-2-thiazolyl)-2, 5-diphenyl-2H-tetrazolium bromide, or MTT, assay, colony formation assays, BrdUrd labelling, immunofluorescence, anchorage-independent growth ability assay and flow cytometry analysis were used to test the effect of miR-27-3p. Luciferase assays were added to verify the direct relationship between miR-27-3p and the predicted target gene inhibitor of growth family member 5 (ING5). RESULTS The expression of miR-27-3p was significantly increased in examined osteosarcoma cell lines compared with that in normal osteocyte cell lines. Up-regulation of miR-27-3p significantly accelerated osteosarcoma cell growth via promoting G1-S transition. In addition, the opposite result was observed in miR-27-3p-down-regulated cells. Up-regulation of ING5 significantly attenuated the miR-27-3p-induced proliferation in osteosarcoma cells. CONCLUSIONS These data suggested that miR-27-3p could promote the G1-S phase transition that leads to proliferation by down-regulating the expression of ING5 in osteosarcoma.
Collapse
Affiliation(s)
- Pei Ye
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Xueping Ke
- Department of Gastroenterology, the Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510175, China
| | - Xuehui Zang
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Hui Sun
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Zhixing Dong
- Department of Radiology, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Jun Lin
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Lihui Wang
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Wenzhou Liu
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Guiqiang Miao
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Yongtao Tan
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Weilai Tong
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Haichang Xiao
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Lihua Gao
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China.
| |
Collapse
|
16
|
Abudesimu A, Adi D, Siti D, Ma X, Liu F, Xie X, Yang Y, Li X, Chen B, Ma Y, Fu Z. Association of lipid metabolism relevant gene FBXW7 polymorphism with coronary artery disease in Uygur Chinese population in Xinjiang, China: a case-control. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:11179-11187. [PMID: 31966468 PMCID: PMC6965840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/18/2017] [Indexed: 06/10/2023]
Abstract
BACKGROUND Hyperlipidemia is a major risk factor for coronary artery disease (CAD). As F-box and WD repeat domain-containing 7 (FBXW7) gene is an important regulating factor for lipid metabolism, the aim of the present study is to assess the association between human FBXW7 gene polymorphisms and CAD among Han Chinese and Uygur Chinese populations in Xinjiang, China. METHODS A total of 1,312 Han Chinese (650 CAD patients and 662 controls) and 834 Uygur Chinese (414 CAD patients and 420 controls) were enrolled in this case-control study. Three single nucleotide polymorphisms (SNPs) rs2255137 T>C, rs2292743 A>T, rs35311955 G>C of FBXW7 were selected and genotyped using the improved multiplex ligase detection reaction (iMLDR) method. RESULTS We found that the rs2255137 CC genotype was very common in the CAD patients compared with the control subjects in the Uygur Chinese populations. After adjustments for several confounders: age, gender, smoking, drinking, hypertension, diabetes, TG, TC, HDL-C and LDL-C, this association remained significant. Furthermore, we investigated the relationships between rs2255137 genotypes and the circulating serum lipid levels and found that people carrying the C allele of rs2255137 may have higher serum lipid levels in the Uygur Chinese populations. CONCLUSION Our results indicate that rs2255137 in FBXW7 gene is associated with CAD in the Uygur Chinese population in China.
Collapse
Affiliation(s)
- Asiya Abudesimu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| | - Dilare Adi
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| | - Dilixiati Siti
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| | - Xiang Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| | - Fen Liu
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| | - Xiang Xie
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| | - Yining Yang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| | - Xiaomei Li
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| | - Bangdang Chen
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| | - Yitong Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| | - Zhenyan Fu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, PR China
| |
Collapse
|
17
|
Zheng HC, Zhao S, Song Y, Ding XQ. The roles of ING5 expression in ovarian carcinogenesis and subsequent progression: a target of gene therapy. Oncotarget 2017; 8:103449-103464. [PMID: 29262575 PMCID: PMC5732741 DOI: 10.18632/oncotarget.21968] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/21/2017] [Indexed: 12/28/2022] Open
Abstract
Here, we found that ING5 overexpression suppressed cell viability, glucose metabolism, migration, invasion and epithelial-mesenchymal transition, and induced cell arrest, apoptosis, senescence, autophagy and fat accumulation in ovarian cancer cells. ING5-mediated chemoresistance was positively linked to apoptotic resistance and chemoresistance-related gene expression. ING5 overexpression suppressed tumor growth of ovarian cancer by decreasing proliferation, and inducing apoptosis and autophagy. ING5 mRNA level was lower in ovarian cancer than normal ovary, and borderline than benign tumors (p < 0.05), and negatively correlated with vascular invasion, lymphatic invasion and FIGO staging of ovarian cancer (p < 0.05). ING5 protein was less expressed in primary cancer than normal ovary (p < 0.05). There was a negative correlation between ING5 mRNA expression and the overall or progression-free survival time of the cancer patients with Grade 2, Grade 3, and stage I cancer (p < 0.05). Immunohistochemically, ING5 was less expressed in serous and mucinous adenocarcinoma than miscellaneous subtypes, and positively correlated with dedifferentiation and ki-67 expression of ovarian cancer (p < 0.05). These data suggested that down-regulated ING5 expression might be involved in ovarian carcinogenesis possibly by suppressing aggressive phenotypes, including proliferation, tumor growth, migration, invasion, and anti-apoptosis.
Collapse
Affiliation(s)
- Hua-Chuan Zheng
- Department of Experimental Oncology and Animal Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Shuang Zhao
- Department of Experimental Oncology and Animal Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yang Song
- Department of Pathology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Xiao-Qing Ding
- Department of Experimental Oncology and Animal Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
18
|
Wang F, Wang AY, Chesnelong C, Yang Y, Nabbi A, Thalappilly S, Alekseev V, Riabowol K. ING5 activity in self-renewal of glioblastoma stem cells via calcium and follicle stimulating hormone pathways. Oncogene 2017; 37:286-301. [PMID: 28925404 PMCID: PMC5799773 DOI: 10.1038/onc.2017.324] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/10/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022]
Abstract
Stem cell-like brain tumor initiating cells (BTICs) cause recurrence of glioblastomas, with BTIC 'stemness' affected by epigenetic mechanisms. The ING family of epigenetic regulators (ING1-5) function by targeting histone acetyltransferase (HAT) or histone deacetylase complexes to the H3K4me3 mark to alter histone acetylation and subsequently, gene expression. Here we find that ectopic expression of ING5, the targeting subunit of HBO1, MOZ and MORF HAT complexes increases expression of the Oct4, Olig2 and Nestin stem cell markers, promotes self-renewal, prevents lineage differentiation and increases stem cell pools in BTIC populations. This activity requires the plant homeodomain region of ING5 that interacts specifically with the H3K4me3 mark. ING5 also enhances PI3K/AKT and MEK/ERK activity to sustain self-renewal of BTICs over serial passage of stem cell-like spheres. ING5 exerts these effects by activating transcription of calcium channel and follicle stimulating hormone pathway genes. In silico analyses of The Cancer Genome Atlas data suggest that ING5 is a positive regulator of BTIC stemness, whose expression negatively correlates with patient prognosis, especially in the Proneural and Classical subtypes, and in tumors with low SOX2 expression. These data suggest that altering histone acetylation status and signaling pathways induced by ING5 may provide useful clinical strategies to target tumor resistance and recurrence in glioblastoma.
Collapse
Affiliation(s)
- F Wang
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - A Y Wang
- Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - C Chesnelong
- Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Y Yang
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - A Nabbi
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - S Thalappilly
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - V Alekseev
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - K Riabowol
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|