1
|
Zhang H, Li Y, Lu H. Correlation of BUB1 and BUB1B with the development and prognosis of endometrial cancer. Sci Rep 2024; 14:17084. [PMID: 39048649 PMCID: PMC11269704 DOI: 10.1038/s41598-024-67528-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
This study aimed to evaluate the expression and clinical significance of budding uninhibited by benzimidazole 1 (BUB1) and BUB1 mitotic checkpoint serine/threonine kinase B (BUB1B) in endometrial carcinoma (EC). BUB1 and BUBIB expressions were evaluated by bioinformatics. Protein expression, clinical features, prognosis and immune cell infiltration were explored in 20 EC tumors. siRNA was used to evaluate BUB1 and BUBIB function in EC cells. BUB1 and BUBIB were highly expressed in 26 cancers. BUB1 was associated with overall survival (OS) in eight cancers and disease-free survival in ten; BUB1B was associated with OS in nine cancers and DFS in eleven. BUB1 and BUBIB exhibited high frequencies of gene changes (mainly mutations, > 5%) in cancer. BUB1 was negatively correlated and BUB1B was positively correlated with cancer-associated fibroblasts and endothelial cell infiltration. BUB1 and BUBIB knockdown decreased migration and invasion in EC cells. High BUB1 expression correlated with tumor malignant phenotypes (P < 0.05). High BUB1 mRNA expression reduced OS (P = 0.00036) and recurrence-free survival (P = 0.0011). High BUB1B mRNA expression reduced OS (P = 0.0024). BUB1/BUB1B correlated with activated CD8 + T and CD4 + T cell infiltration. BUB1 and BUBIB are highly expressed and correlated with clinicopathological characteristics in EC. BUB1 and BUBIB are potential prognosis markers and immunotherapy targets.
Collapse
Affiliation(s)
- Huicong Zhang
- Clinical Medicinal College of Dali University, Dali City, 671000, Yunnan Province, China
| | - Yuhao Li
- West China School of Basic Medical Sciences and Forensic Medicine,, Sichuan University, 610041, Chengdu, China
| | - Huixia Lu
- Clinical Medicinal College of Dali University, Dali City, 671000, Yunnan Province, China.
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, 671000, Yunnan Province, China.
| |
Collapse
|
2
|
Hosea R, Duan W, Meliala ITS, Li W, Wei M, Hillary S, Zhao H, Miyagishi M, Wu S, Kasim V. YY2/BUB3 Axis promotes SAC Hyperactivation and Inhibits Colorectal Cancer Progression via Regulating Chromosomal Instability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308690. [PMID: 38682484 PMCID: PMC11234461 DOI: 10.1002/advs.202308690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/08/2024] [Indexed: 05/01/2024]
Abstract
Spindle assembly checkpoint (SAC) is a crucial safeguard mechanism of mitosis fidelity that ensures equal division of duplicated chromosomes to the two progeny cells. Impaired SAC can lead to chromosomal instability (CIN), a well-recognized hallmark of cancer that facilitates tumor progression; paradoxically, high CIN levels are associated with better therapeutic response and prognosis. However, the mechanism by which CIN determines tumor cell survival and therapeutic response remains poorly understood. Here, using a cross-omics approach, YY2 is identified as a mitotic regulator that promotes SAC activity by activating the transcription of budding uninhibited by benzimidazole 3 (BUB3), a component of SAC. While both conditions induce CIN, a defect in YY2/SAC activity enhances mitosis and tumor growth. Meanwhile, hyperactivation of SAC mediated by YY2/BUB3 triggers a delay in mitosis and suppresses growth. Furthermore, it is revealed that YY2/BUB3-mediated excessive CIN causes higher cell death rates and drug sensitivity, whereas residual tumor cells that survived DNA damage-based therapy have moderate CIN and increased drug resistance. These results provide insights into the role of SAC activity and CIN levels in influencing tumor cell survival and drug response, as well as suggest a novel anti-tumor therapeutic strategy that combines SAC activity modulators and DNA-damage agents.
Collapse
Affiliation(s)
- Rendy Hosea
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Wei Duan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Ian Timothy Sembiring Meliala
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Wenfang Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Mankun Wei
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Sharon Hillary
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Hezhao Zhao
- Department of Gastrointestinal Surgery, Chongqing University Cancer HospitalChongqing UniversityChongqing400030P. R. China
| | - Makoto Miyagishi
- Life Science Innovation, School of Integrative and Global MajorsUniversity of TsukubaTsukubaIbaraki305‐0006Japan
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer HospitalChongqing UniversityChongqing400030P. R. China
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer HospitalChongqing UniversityChongqing400030P. R. China
| |
Collapse
|
3
|
Zheng Q, Luo Z, Xu M, Ye S, Lei Y, Xi Y. HMGA1 and FOXM1 Cooperate to Promote G2/M Cell Cycle Progression in Cancer Cells. Life (Basel) 2023; 13:life13051225. [PMID: 37240870 DOI: 10.3390/life13051225] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/14/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
HMGA1 is a chromatin-binding protein and performs its biological function by remodeling chromatin structure or recruiting other transcription factors. However, the role of abnormally high level of HMGA1 in cancer cells and its regulatory mechanism still require further investigation. In this study, we performed a prognostic analysis and showed that high level of either HMGA1 or FOXM1 was associated with poor prognosis in various cancers based on the TCGA database. Furthermore, the expression pattern of HMGA1 and FOXM1 showed a significant strong positive correlation in most type of cancers, especially lung adenocarcinoma, pancreatic cancer and liver cancer. Further analysis of the biological effects of their high correlation in cancers suggested that cell cycle was the most significant related pathway commonly regulated by HMGA1 and FOXM1. After knockdown of HMGA1 and FOXM1 by specific siRNAs, an obvious increased G2/M phase was observed in the siHMGA1 and siFOXM1 groups compared to the siNC group. The expression levels of key G2/M phase regulatory genes PLK1 and CCNB1 were significantly downregulated. Importantly, HMGA1 and FOXM1 were identified to form a protein complex and co-located in the nucleus based on co-immunoprecipitation and immunofluorescence staining, respectively. Thus, our results provide the basic evidence that HMGA1 and FOXM1 cooperatively accelerate cell cycle progression by up-regulating PLK1 and CCNB1 to promote cancer cell proliferation.
Collapse
Affiliation(s)
- Qingfang Zheng
- Institute of Biochemistry and Molecular Biology, Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Ziyang Luo
- Institute of Biochemistry and Molecular Biology, Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Mingjun Xu
- Institute of Biochemistry and Molecular Biology, Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Shazhou Ye
- Institute of Biochemistry and Molecular Biology, Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yuxin Lei
- Institute of Biochemistry and Molecular Biology, Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yang Xi
- Institute of Biochemistry and Molecular Biology, Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China
| |
Collapse
|
4
|
Wang L, Zhang J, Xia M, Liu C, Zu X, Zhong J. High Mobility Group A1 (HMGA1): Structure, Biological Function, and Therapeutic Potential. Int J Biol Sci 2022; 18:4414-4431. [PMID: 35864955 PMCID: PMC9295051 DOI: 10.7150/ijbs.72952] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022] Open
Abstract
High mobility group A1 (HMGA1) is a nonhistone chromatin structural protein characterized by no transcriptional activity. It mainly plays a regulatory role by modifying the structure of DNA. A large number of studies have confirmed that HMGA1 regulates genes related to tumours in the reproductive system, digestive system, urinary system and haematopoietic system. HMGA1 is rare in adult cells and increases in highly proliferative cells such as embryos. After being stimulated by external factors, it will produce effects through the Wnt/β-catenin, PI3K/Akt, Hippo and MEK/ERK pathways. In addition, HMGA1 also affects the ageing, apoptosis, autophagy and chemotherapy resistance of cancer cells, which are linked to tumorigenesis. In this review, we summarize the mechanisms of HMGA1 in cancer progression and discuss the potential clinical application of targeted HMGA1 therapy, indicating that targeted HMGA1 is of great significance in the diagnosis and treatment of malignancy.
Collapse
Affiliation(s)
- Lu Wang
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Ji Zhang
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
| | - Min Xia
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Chang Liu
- Department of Endocrinology and Metabolism, The First People's Hospital of Chenzhou, First School of Clinical Medicine, University of Southern Medical, Guangzhou 510515, Guangdong, China
| | - Xuyu Zu
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jing Zhong
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| |
Collapse
|
5
|
De Martino M, Esposito F, Fusco A. Critical role of the high mobility group A proteins in hematological malignancies. Hematol Oncol 2021; 40:2-10. [PMID: 34637548 PMCID: PMC9293314 DOI: 10.1002/hon.2934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 12/17/2022]
Abstract
The high mobility group A (HMGA) protein family is composed of three non‐histone chromatin remodeling proteins that act as architectural transcriptional factors. Indeed, although HMGA proteins lack transcriptional activity per se, they bind the minor groove of DNA at AT‐rich sequences, and, interacting with the transcription machinery, are able to modify chromatin modeling, thus regulating the expression of several genes. HMGA proteins have been deeply involved in embryogenesis process, and a large volume of studies has pointed out their key role in human cancer. Here, we review the studies on the role of the HMGA proteins in human hematological malignancies: they are overexpressed in most of the cases and their expression correlates with a reduced survival. In some cases, such as in acute lymphoblastic leukemia and acute myelogenous leukemia, HMGA2 gene rearrangements have been also described. Finally, recent studies evidence a synergism between HMGA and EZH2 in diffuse B‐cell lymphomas, suggesting an innovative therapy for this disease based on the inhibition of the function of both these proteins.
Collapse
Affiliation(s)
- Marco De Martino
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), National Research Council (CNR), Institute for Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", University of Naples "Federico II", Naples, Italy.,Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesco Esposito
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), National Research Council (CNR), Institute for Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", University of Naples "Federico II", Naples, Italy
| | - Alfredo Fusco
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), National Research Council (CNR), Institute for Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", University of Naples "Federico II", Naples, Italy
| |
Collapse
|
6
|
Papantonis A. HMGs as rheostats of chromosomal structure and cell proliferation. Trends Genet 2021; 37:986-994. [PMID: 34311989 DOI: 10.1016/j.tig.2021.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/30/2021] [Accepted: 07/03/2021] [Indexed: 11/18/2022]
Abstract
High mobility group proteins (HMGs) are the most abundant nuclear proteins next to histones and are robustly expressed across tissues and organs. HMGs can uniquely bend or bind distorted DNA, and are central to such processes as transcription, recombination, and DNA repair. However, their dynamic association with chromatin renders capturing HMGs on chromosomes challenging. Recent work has changed this and now implicates these factors in spatial genome organization. Here, I revisit older and review recent literature to describe how HMGs rewire spatial chromatin interactions to sustain homeostasis or promote cellular aging. I propose a 'rheostat' model to explain how HMG-box proteins (HMGBs), and to some extent HMG A proteins (HMGAs), may control cellular aging and, likely, cancer progression.
Collapse
Affiliation(s)
- Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany.
| |
Collapse
|
7
|
De Martino M, Nicolau-Neto P, Ribeiro Pinto LF, Traverse-Glehen A, Bachy E, Gigantino V, De Cecio R, Bertoni F, Chieffi P, Fusco A, Esposito F. HMGA1 induces EZH2 overexpression in human B-cell lymphomas. Am J Cancer Res 2021; 11:2174-2187. [PMID: 34094676 PMCID: PMC8167683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/03/2021] [Indexed: 06/12/2023] Open
Abstract
EZH2 is an enzymatic subunit of PRC2, an epigenetic regulator that triggers the methylation of the histone H3 lysine 27 silencing the transcription of several genes. EZH2 has a critical role in cancer progression, since its overexpression has been associated with increased cancer cell invasiveness, drug resistance and poor patient survival. However, the mechanisms accounting for EZH2 overexpression in cancer remain still unclear. Intriguingly, also HMGA protein overexpression is a feature of many human malignancies and correlates with the presence of metastases and a poor outcome. The HMGA proteins, including HMGA1 and HMGA2, belong to the architectural transcription factors that play a key role in the organization of chromatin structure. Here, we report a statistically significant correlation between HMGA1 and EZH2 expression in human lymphomas. We demonstrate that HMGA1 is able to bind EZH2 promoter and induce its activity. Consistently, silencing of HMGA1 expression results in the downregulation of the EZH2 levels leading to a decreased proliferation and migration rate of human lymphoma cell lines. Therefore, these data identify HMGA1 as an EZH2 activator, suggesting a novel molecular mechanism contributing to EZH2 overexpression in human malignancies and a synergism of these proteins in cancer progression.
Collapse
Affiliation(s)
- Marco De Martino
- Institute of Endocrinology and Experimental Oncology-CNR c/o Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”Naples, Italy
| | - Pedro Nicolau-Neto
- Molecular Carcinogenesis Program, National Cancer Institute-INCARua Andre Cavalcanti 37, Rio de Janeiro 20231-050, Brazil
| | - Luis Felipe Ribeiro Pinto
- Molecular Carcinogenesis Program, National Cancer Institute-INCARua Andre Cavalcanti 37, Rio de Janeiro 20231-050, Brazil
- Department of Biochemistry, Roberto Alcantara Gomes Biology Institute, State University of Rio de JaneiroRio de Janeiro 20551-030, Brazil
| | - Alexandra Traverse-Glehen
- Hospices Civils de Lyon, Department of Pathological AnatomyLyon, France
- Claude Bernard Lyon 1 UniversityLyon, France
| | - Emmanuel Bachy
- Claude Bernard Lyon 1 UniversityLyon, France
- Department of Hematology, Hospices Civils de Lyon, Lyon Sud HospitalPierre-Bénite, France
| | - Vincenzo Gigantino
- Pathology Unit, National Cancer Institute, IRCCS, Pascale FoundationNaples, Italy
| | - Rossella De Cecio
- Pathology Unit, National Cancer Institute, IRCCS, Pascale FoundationNaples, Italy
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USIBellinzona, Switzerland
- Oncology Institute of Southern SwitzerlandBellinzona, Switzerland
| | - Paolo Chieffi
- Department of Psychology, University of Campania “L. Vanvitelli”Caserta, Italy
| | - Alfredo Fusco
- Institute of Endocrinology and Experimental Oncology-CNR c/o Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”Naples, Italy
| | - Francesco Esposito
- Institute of Endocrinology and Experimental Oncology-CNR c/o Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”Naples, Italy
| |
Collapse
|
8
|
Baumann C, Zhang X, De La Fuente R. Loss of CBX2 induces genome instability and senescence-associated chromosomal rearrangements. J Cell Biol 2021; 219:152063. [PMID: 32870972 PMCID: PMC7594495 DOI: 10.1083/jcb.201910149] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 06/08/2020] [Accepted: 08/02/2020] [Indexed: 01/05/2023] Open
Abstract
The polycomb group protein CBX2 is an important epigenetic reader involved in cell proliferation and differentiation. While CBX2 overexpression occurs in a wide range of human tumors, targeted deletion results in homeotic transformation, proliferative defects, and premature senescence. However, its cellular function(s) and whether it plays a role in maintenance of genome stability remain to be determined. Here, we demonstrate that loss of CBX2 in mouse fibroblasts induces abnormal large-scale chromatin structure and chromosome instability. Integrative transcriptome analysis and ATAC-seq revealed a significant dysregulation of transcripts involved in DNA repair, chromocenter formation, and tumorigenesis in addition to changes in chromatin accessibility of genes involved in lateral sclerosis, basal transcription factors, and folate metabolism. Notably, Cbx2−/− cells exhibit prominent decondensation of satellite DNA sequences at metaphase and increased sister chromatid recombination events leading to rampant chromosome instability. The presence of extensive centromere and telomere defects suggests a prominent role for CBX2 in heterochromatin homeostasis and the regulation of nuclear architecture.
Collapse
Affiliation(s)
- Claudia Baumann
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA.,Regenerative Bioscience Center, University of Georgia, Athens, GA
| | - Xiangyu Zhang
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA.,Regenerative Bioscience Center, University of Georgia, Athens, GA
| | - Rabindranath De La Fuente
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA.,Regenerative Bioscience Center, University of Georgia, Athens, GA
| |
Collapse
|
9
|
Meireles Da Costa N, Palumbo A, De Martino M, Fusco A, Ribeiro Pinto LF, Nasciutti LE. Interplay between HMGA and TP53 in cell cycle control along tumor progression. Cell Mol Life Sci 2021; 78:817-831. [PMID: 32920697 PMCID: PMC11071717 DOI: 10.1007/s00018-020-03634-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/05/2020] [Accepted: 09/03/2020] [Indexed: 01/27/2023]
Abstract
The high mobility group A (HMGA) proteins are found to be aberrantly expressed in several tumors. Studies (in vitro and in vivo) have shown that HMGA protein overexpression has a causative role in carcinogenesis process. HMGA proteins regulate cell cycle progression through distinct mechanisms which strongly influence its normal dynamics along malignant transformation. Tumor protein p53 (TP53) is the most frequently altered gene in cancer. The loss of its activity is recognized as the fall of a barrier that enables neoplastic transformation. Among the different functions, TP53 signaling pathway is tightly involved in control of cell cycle, with cell cycle arrest being the main biological outcome observed upon p53 activation, which prevents accumulation of damaged DNA, as well as genomic instability. Therefore, the interaction and opposing effects of HMGA and p53 proteins on regulation of cell cycle in normal and tumor cells are discussed in this review. HMGA proteins and p53 may reciprocally regulate the expression and/or activity of each other, leading to the counteraction of their regulation mechanisms at different stages of the cell cycle. The existence of a functional crosstalk between these proteins in the control of cell cycle could open the possibility of targeting HMGA and p53 in combination with other therapeutic strategies, particularly those that target cell cycle regulation, to improve the management and prognosis of cancer patients.
Collapse
Affiliation(s)
- Nathalia Meireles Da Costa
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer-INCA, Rua André Cavalcanti, 37-6th floor-Centro, 20231-050, Rio de Janeiro, RJ, Brazil.
| | - Antonio Palumbo
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Prédio de Ciências da Saúde-Cidade Universitária, Ilha do Fundão, A. Carlos Chagas, 373-Bloco F, Sala 26, 21941-902, Rio de Janeiro, RJ, Brazil
| | - Marco De Martino
- Istituto di Endocrinologia e Oncologia Sperimentale-CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Alfredo Fusco
- Istituto di Endocrinologia e Oncologia Sperimentale-CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Luis Felipe Ribeiro Pinto
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer-INCA, Rua André Cavalcanti, 37-6th floor-Centro, 20231-050, Rio de Janeiro, RJ, Brazil
| | - Luiz Eurico Nasciutti
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Prédio de Ciências da Saúde-Cidade Universitária, Ilha do Fundão, A. Carlos Chagas, 373-Bloco F, Sala 26, 21941-902, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
10
|
Double knock-out of Hmga1 and Hipk2 genes causes perinatal death associated to respiratory distress and thyroid abnormalities in mice. Cell Death Dis 2019; 10:747. [PMID: 31582725 PMCID: PMC6776533 DOI: 10.1038/s41419-019-1975-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 09/03/2019] [Accepted: 09/12/2019] [Indexed: 12/02/2022]
Abstract
The serine–threonine kinase homeodomain-interacting protein kinase 2 (HIPK2) modulates important cellular functions during development, acting as a signal integrator of a wide variety of stress signals, and as a regulator of transcription factors and cofactors. We have previously demonstrated that HIPK2 binds and phosphorylates High-Mobility Group A1 (HMGA1), an architectural chromatinic protein ubiquitously expressed in embryonic tissues, decreasing its binding affinity to DNA. To better define the functional role of HIPK2 and HMGA1 interaction in vivo, we generated mice in which both genes are disrupted. About 50% of these Hmga1/Hipk2 double knock-out (DKO) mice die within 12 h of life (P1) for respiratory failure. The DKO mice present an altered lung morphology, likely owing to a drastic reduction in the expression of surfactant proteins, that are required for lung development. Consistently, we report that both HMGA1 and HIPK2 proteins positively regulate the transcriptional activity of the genes encoding the surfactant proteins. Moreover, these mice display an altered expression of thyroid differentiation markers, reasonably because of a drastic reduction in the expression of the thyroid-specific transcription factors PAX8 and FOXE1, which we demonstrate here to be positively regulated by HMGA1 and HIPK2. Therefore, these data indicate a critical role of HIPK2/HMGA1 cooperation in lung and thyroid development and function, suggesting the potential involvement of their impairment in the pathogenesis of human lung and thyroid diseases.
Collapse
|
11
|
Puca F, Tosti N, Federico A, Kuzay Y, Pepe A, Morlando S, Savarese T, D’Alessio F, Colamaio M, Sarnataro D, Ziberi S, De Martino M, Fusco A, Battista S. HMGA1 negatively regulates NUMB expression at transcriptional and post transcriptional level in glioblastoma stem cells. Cell Cycle 2019; 18:1446-1457. [PMID: 31116627 PMCID: PMC6592240 DOI: 10.1080/15384101.2019.1618541] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/02/2019] [Accepted: 05/08/2019] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is a lethal, fast-growing brain cancer, affecting 2-3 per 100,000 adults per year. It arises from multipotent neural stem cells which have reduced their ability to divide asymmetrically and hence divide symmetrically, generating increasing number of cancer stem cells, fostering tumor growth. We have previously demonstrated that the architectural transcription factor HMGA1 is highly expressed in brain tumor stem cells (BTSCs) and that its silencing increases stem cell quiescence, reduces self-renewal and sphere-forming efficiency in serial passages, suggesting a shift from symmetric to asymmetric division. Since NUMB expression is fundamental for the fulfillment of asymmetric division in stem cells, and is lost or reduced in many tumors, including GBM, we have investigated the ability of HMGA1 to regulate NUMB expression. Here, we show that HMGA1 negatively regulates NUMB expression at transcriptional level, by binding its promoter and counteracting c/EBP-β and at posttranscriptional level, by regulating the expression of MSI1 and of miR-146a. Finally, we report that HMGA1 knockdown-induced NUMB upregulation leads to the downregulation of the NOTCH1 pathway. Therefore, the data reported here indicate that HMGA1 negatively regulates NUMB expression in BTSCs, further supporting HMGA1 targeting as innovative and effective anti-cancer therapy.
Collapse
Affiliation(s)
- Francesca Puca
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Nadia Tosti
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Antonella Federico
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Yalçın Kuzay
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Anna Pepe
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Sonia Morlando
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Teresa Savarese
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Federica D’Alessio
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Marianna Colamaio
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Daniela Sarnataro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
- Dynamic Imaging and Microscopy Facility, CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Sihana Ziberi
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche dell’Università “G. d’Annunzio” di Chieti, Chieti, Italy
| | - Marco De Martino
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Alfredo Fusco
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Sabrina Battista
- Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| |
Collapse
|
12
|
Portovedo S, Gaido N, de Almeida Nunes B, Nascimento AG, Rocha A, Magalhães M, Nascimento GC, Pires de Carvalho D, Soares P, Takiya C, Faria MDS, Miranda-Alves L. Differential Expression of HMGA1 and HMGA2 in pituitary neuroendocrine tumors. Mol Cell Endocrinol 2019; 490:80-87. [PMID: 30999005 DOI: 10.1016/j.mce.2019.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/05/2019] [Accepted: 04/12/2019] [Indexed: 02/06/2023]
Abstract
Defining biomarkers for invasive pituitary neuroendocrine tumors (PitNETs) is highly desirable. The high mobility group A (HMGA) proteins are among the most widely expressed cancer-associated proteins. Indeed, their overexpression is a frequent feature of human malignancies, including PitNETs. We show that nonfunctioning PitNETs (NF-PitNETs) express significantly higher levels of HMGA1 than somatotropinomas (GHs) and corticotropinomas (ACTHs). Furthermore, HMGA2 expression was detected only in NF-PitNETs and was significantly higher in larger tumors than in smaller tumors. HMGA expression analysis generally focuses on nuclear staining. Here, cytoplasmic HMGA staining was also found. PitNETs displayed strong nuclear HMGA1 and strong cytoplasmic HMGA2 immunoreactivity. Interestingly, the HMGA1 and HMGA2 nuclear expression levels were significantly higher in invasive adenomas than in noninvasive adenomas. The highest levels of nuclear HMGA2 were found in GHs. In conclusion, we show that overexpression of nuclear HMGA proteins could be a potential biomarker of invasive PitNETs, particularly HMGA2 for GHs. HMGA2 might be a reliable biomarker for NF-PitNETs.
Collapse
Affiliation(s)
- Sérgio Portovedo
- Laboratory of Experimental Endocrinology - LEEx, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Graduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Brazil
| | - Nadja Gaido
- Service of Endocrinology, President Dutra Hospital of the Federal University of Maranhão and Clinical Research Center of the President Dutra Hospital of the Federal University of Maranhão, Brazil
| | - Bruno de Almeida Nunes
- Laboratory of Experimental Endocrinology - LEEx, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Graduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Brazil
| | - Ana Giselia Nascimento
- Service of Pathology, President Dutra Hospital of the Federal University of Maranhão, Brazil
| | - Allysson Rocha
- Department of Radiology and Diagnostic Imaging, President Dutra Hospital, Federal University of Maranhão, Brazil
| | - Marcelo Magalhães
- Service of Endocrinology, President Dutra Hospital of the Federal University of Maranhão and Clinical Research Center of the President Dutra Hospital of the Federal University of Maranhão, Brazil
| | - Gilvan Cortes Nascimento
- Service of Endocrinology, President Dutra Hospital of the Federal University of Maranhão and Clinical Research Center of the President Dutra Hospital of the Federal University of Maranhão, Brazil
| | - Denise Pires de Carvalho
- Graduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Brazil; Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Brazil
| | - Paula Soares
- Laboratory of Experimental Endocrinology - LEEx, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Graduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Brazil; Institute for Research and Innovation in Health (I3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) - Cancer Signaling & Metabolism, Portugal; Department of Pathology, Faculty of Medicine, University of Porto, Portugal
| | - Christina Takiya
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Brazil
| | - Manuel Dos Santos Faria
- Laboratory of Experimental Endocrinology - LEEx, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Service of Endocrinology, President Dutra Hospital of the Federal University of Maranhão and Clinical Research Center of the President Dutra Hospital of the Federal University of Maranhão, Brazil
| | - Leandro Miranda-Alves
- Laboratory of Experimental Endocrinology - LEEx, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Graduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Brazil; Graduate Program in Pharmacology and Medicinal Chemistry, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
Wang Y, Hu L, Zheng Y, Guo L. HMGA1 in cancer: Cancer classification by location. J Cell Mol Med 2019; 23:2293-2302. [PMID: 30614613 PMCID: PMC6433663 DOI: 10.1111/jcmm.14082] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 07/19/2018] [Accepted: 11/16/2018] [Indexed: 12/23/2022] Open
Abstract
The high mobility group A1 (HMGA1) gene plays an important role in numerous malignant cancers. HMGA1 is an oncofoetal gene, and we have a certain understanding of the biological function of HMGA1 based on its activities in various neoplasms. As an architectural transcription factor, HMGA1 remodels the chromatin structure and promotes the interaction between transcriptional regulatory proteins and DNA in different cancers. Through analysis of the molecular mechanism of HMGA1 and clinical studies, emerging evidence indicates that HMGA1 promotes the occurrence and metastasis of cancer. Within a similar location or the same genetic background, the function and role of HMGA1 may have certain similarities. In this paper, to characterize HMGA1 comprehensively, research on various types of tumours is discussed to further understanding of the function and mechanism of HMGA1. The findings provide a more reliable basis for classifying HMGA1 function according to the tumour location. In this review, we summarize recent studies related to HMGA1, including its structure and oncogenic properties, its major functions in each cancer, its upstream and downstream regulation associated with the tumourigenesis and metastasis of cancer, and its potential as a biomarker for clinical diagnosis of cancer.
Collapse
Affiliation(s)
- Yuhong Wang
- The First Affiliated Hospital of Soochow University Department of Pathology, Suzhou, Jiangsu, China
| | - Lin Hu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yushuang Zheng
- The First Affiliated Hospital of Soochow University Department of Pathology, Suzhou, Jiangsu, China
| | - Lingchuan Guo
- The First Affiliated Hospital of Soochow University Department of Pathology, Suzhou, Jiangsu, China
| |
Collapse
|
14
|
Bellato M, De Marchi D, Gualtieri C, Sauta E, Magni P, Macovei A, Pasotti L. A Bioinformatics Approach to Explore MicroRNAs as Tools to Bridge Pathways Between Plants and Animals. Is DNA Damage Response (DDR) a Potential Target Process? FRONTIERS IN PLANT SCIENCE 2019; 10:1535. [PMID: 31850028 PMCID: PMC6901925 DOI: 10.3389/fpls.2019.01535] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 11/04/2019] [Indexed: 05/10/2023]
Abstract
MicroRNAs, highly-conserved small RNAs, act as key regulators of many biological functions in both plants and animals by post-transcriptionally regulating gene expression through interactions with their target mRNAs. The microRNA research is a dynamic field, in which new and unconventional aspects are emerging alongside well-established roles in development and stress adaptation. A recent hypothesis states that miRNAs can be transferred from one species to another and potentially target genes across distant species. Here, we propose to look into the trans-kingdom potential of miRNAs as a tool to bridge conserved pathways between plant and human cells. To this aim, a novel multi-faceted bioinformatic analysis pipeline was developed, enabling the investigation of common biological processes and genes targeted in plant and human transcriptome by a set of publicly available Medicago truncatula miRNAs. Multiple datasets, including miRNA, gene, transcript and protein sequences, expression profiles and genetic interactions, were used. Three different strategies were employed, namely a network-based pipeline, an alignment-based pipeline, and a M. truncatula network reconstruction approach, to study functional modules and to evaluate gene/protein similarities among miRNA targets. The results were compared in order to find common features, e.g., microRNAs targeting similar processes. Biological processes like exocytosis and response to viruses were common denominators in the investigated species. Since the involvement of miRNAs in the regulation of DNA damage response (DDR)-associated pathways is barely explored, especially in the plant kingdom, a special attention is given to this aspect. Hereby, miRNAs predicted to target genes involved in DNA repair, recombination and replication, chromatin remodeling, cell cycle and cell death were identified in both plants and humans, paving the way for future interdisciplinary advancements.
Collapse
Affiliation(s)
- Massimo Bellato
- Laboratory of Bioinformatics, Mathematical Modelling, and Synthetic Biology, Department of Electrical, Computer and Biomedical Engineering—Centre for Health Technology, University of Pavia, Pavia, Italy
| | - Davide De Marchi
- Laboratory of Bioinformatics, Mathematical Modelling, and Synthetic Biology, Department of Electrical, Computer and Biomedical Engineering—Centre for Health Technology, University of Pavia, Pavia, Italy
| | - Carla Gualtieri
- Plant Biotechnology Laboratory, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Elisabetta Sauta
- Laboratory of Bioinformatics, Mathematical Modelling, and Synthetic Biology, Department of Electrical, Computer and Biomedical Engineering—Centre for Health Technology, University of Pavia, Pavia, Italy
| | - Paolo Magni
- Laboratory of Bioinformatics, Mathematical Modelling, and Synthetic Biology, Department of Electrical, Computer and Biomedical Engineering—Centre for Health Technology, University of Pavia, Pavia, Italy
| | - Anca Macovei
- Plant Biotechnology Laboratory, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
- *Correspondence: Anca Macovei, ; Lorenzo Pasotti,
| | - Lorenzo Pasotti
- Laboratory of Bioinformatics, Mathematical Modelling, and Synthetic Biology, Department of Electrical, Computer and Biomedical Engineering—Centre for Health Technology, University of Pavia, Pavia, Italy
- *Correspondence: Anca Macovei, ; Lorenzo Pasotti,
| |
Collapse
|
15
|
Thompson LL, Jeusset LMP, Lepage CC, McManus KJ. Evolving Therapeutic Strategies to Exploit Chromosome Instability in Cancer. Cancers (Basel) 2017; 9:cancers9110151. [PMID: 29104272 PMCID: PMC5704169 DOI: 10.3390/cancers9110151] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 12/12/2022] Open
Abstract
Cancer is a devastating disease that claims over 8 million lives each year. Understanding the molecular etiology of the disease is critical to identify and develop new therapeutic strategies and targets. Chromosome instability (CIN) is an abnormal phenotype, characterized by progressive numerical and/or structural chromosomal changes, which is observed in virtually all cancer types. CIN generates intratumoral heterogeneity, drives cancer development, and promotes metastatic progression, and thus, it is associated with highly aggressive, drug-resistant tumors and poor patient prognosis. As CIN is observed in both primary and metastatic lesions, innovative strategies that exploit CIN may offer therapeutic benefits and better outcomes for cancer patients. Unfortunately, exploiting CIN remains a significant challenge, as the aberrant mechanisms driving CIN and their causative roles in cancer have yet to be fully elucidated. The development and utilization of CIN-exploiting therapies is further complicated by the associated risks for off-target effects and secondary cancers. Accordingly, this review will assess the strengths and limitations of current CIN-exploiting therapies, and discuss emerging strategies designed to overcome these challenges to improve outcomes and survival for patients diagnosed with cancer.
Collapse
Affiliation(s)
- Laura L Thompson
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada.
| | - Lucile M-P Jeusset
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada.
| | - Chloe C Lepage
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada.
| | - Kirk J McManus
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada.
| |
Collapse
|
16
|
Conte A, Paladino S, Bianco G, Fasano D, Gerlini R, Tornincasa M, Renna M, Fusco A, Tramontano D, Pierantoni GM. High mobility group A1 protein modulates autophagy in cancer cells. Cell Death Differ 2017; 24:1948-1962. [PMID: 28777374 PMCID: PMC5635219 DOI: 10.1038/cdd.2017.117] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 06/01/2017] [Accepted: 06/15/2017] [Indexed: 12/11/2022] Open
Abstract
High Mobility Group A1 (HMGA1) is an architectural chromatin protein whose overexpression is a feature of malignant neoplasias with a causal role in cancer initiation and progression. HMGA1 promotes tumor growth by several mechanisms, including increase of cell proliferation and survival, impairment of DNA repair and induction of chromosome instability. Autophagy is a self-degradative process that, by providing energy sources and removing damaged organelles and misfolded proteins, allows cell survival under stress conditions. On the other hand, hyper-activated autophagy can lead to non-apoptotic programmed cell death. Autophagy deregulation is a common feature of cancer cells in which has a complex role, showing either an oncogenic or tumor suppressor activity, depending on cellular context and tumor stage. Here, we report that depletion of HMGA1 perturbs autophagy by different mechanisms. HMGA1-knockdown increases autophagosome formation by constraining the activity of the mTOR pathway, a major regulator of autophagy, and transcriptionally upregulating the autophagy-initiating kinase Unc-51-like kinase 1 (ULK1). Consistently, functional experiments demonstrate that HMGA1 binds ULK1 promoter region and negatively regulates its transcription. On the other hand, the increase in autophagosomes is not associated to a proportionate increase in their maturation. Overall, the effects of HMGA1 depletion on autophagy are associated to a decrease in cell proliferation and ultimately impact on cancer cells viability. Importantly, silencing of ULK1 prevents the effects of HMGA1-knockdown on cellular proliferation, viability and autophagic activity, highlighting how these effects are, at least in part, mediated by ULK1. Interestingly, this phenomenon is not restricted to skin cancer cells, as similar results have been observed also in HeLa cells silenced for HMGA1. Taken together, these results clearly indicate HMGA1 as a key regulator of the autophagic pathway in cancer cells, thus suggesting a novel mechanism through which HMGA1 can contribute to cancer progression.
Collapse
Affiliation(s)
- Andrea Conte
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Gaia Bianco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Dominga Fasano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Raffaele Gerlini
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Mara Tornincasa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Maurizio Renna
- Department of Medical Genetics, Cambridge Institute for Medical Research, Wellcome Trust, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Alfredo Fusco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Donatella Tramontano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Giovanna Maria Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| |
Collapse
|
17
|
Critical role of HMGA proteins in cancer cell chemoresistance. JOURNAL OF MOLECULAR MEDICINE (BERLIN, GERMANY) 2017. [PMID: 28293697 DOI: 10.1007/s00109‐017‐1520‐x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The high-mobility group A (HMGA) proteins are frequently overexpressed in human malignancies and correlate with the presence of metastases and reduced patient survival. Here, we highlight the main studies evidencing a critical role of HMGA in chemoresistance, mainly by activating Akt signaling, impairing p53 activity, and regulating the expression of microRNAs that target genes involved in the susceptibility of cancer cells to antineoplastic agents. Therefore, these studies account for the association of HMGA overexpression with patient poor outcome, indicating the impairment of HMGA as a fascinating perspective for effectively improving cancer therapy.
Collapse
|
18
|
D’Angelo D, Mussnich P, Arra C, Battista S, Fusco A. Critical role of HMGA proteins in cancer cell chemoresistance. J Mol Med (Berl) 2017; 95:353-360. [DOI: 10.1007/s00109-017-1520-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/24/2017] [Accepted: 02/07/2017] [Indexed: 02/03/2023]
|
19
|
Xian L, Reddy KA, Resar LMS. Hmga1 deficiency: "SAC-King" the SAC genes to incite chromosomal instability. Cell Cycle 2017; 16:17-18. [PMID: 27977330 DOI: 10.1080/15384101.2016.1214034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Lingling Xian
- a Department of Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,b Division of Hematology, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Karen A Reddy
- a Department of Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,c Department of Biologic Chemistry , Center for Epigenics, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Linda M S Resar
- a Department of Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,b Division of Hematology, Johns Hopkins University School of Medicine , Baltimore , MD , USA.,d Department of Oncology and Institute for Cellular Engineering, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
20
|
Sumter TF, Xian L, Huso T, Koo M, Chang YT, Almasri TN, Chia L, Inglis C, Reid D, Resar LMS. The High Mobility Group A1 (HMGA1) Transcriptome in Cancer and Development. Curr Mol Med 2016; 16:353-93. [PMID: 26980699 DOI: 10.2174/1566524016666160316152147] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 02/15/2016] [Accepted: 03/10/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND & OBJECTIVES Chromatin structure is the single most important feature that distinguishes a cancer cell from a normal cell histologically. Chromatin remodeling proteins regulate chromatin structure and high mobility group A (HMGA1) proteins are among the most abundant, nonhistone chromatin remodeling proteins found in cancer cells. These proteins include HMGA1a/HMGA1b isoforms, which result from alternatively spliced mRNA. The HMGA1 gene is overexpressed in cancer and high levels portend a poor prognosis in diverse tumors. HMGA1 is also highly expressed during embryogenesis and postnatally in adult stem cells. Overexpression of HMGA1 drives neoplastic transformation in cultured cells, while inhibiting HMGA1 blocks oncogenic and cancer stem cell properties. Hmga1 transgenic mice succumb to aggressive tumors, demonstrating that dysregulated expression of HMGA1 causes cancer in vivo. HMGA1 is also required for reprogramming somatic cells into induced pluripotent stem cells. HMGA1 proteins function as ancillary transcription factors that bend chromatin and recruit other transcription factors to DNA. They induce oncogenic transformation by activating or repressing specific genes involved in this process and an HMGA1 "transcriptome" is emerging. Although prior studies reveal potent oncogenic properties of HMGA1, we are only beginning to understand the molecular mechanisms through which HMGA1 functions. In this review, we summarize the list of putative downstream transcriptional targets regulated by HMGA1. We also briefly discuss studies linking HMGA1 to Alzheimer's disease and type-2 diabetes. CONCLUSION Further elucidation of HMGA1 function should lead to novel therapeutic strategies for cancer and possibly for other diseases associated with aberrant HMGA1 expression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - L M S Resar
- Department of Medicine, Faculty of the Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 1025, Baltimore, MD 21205-2109, USA.
| |
Collapse
|
21
|
Pierantoni GM, Conte A, Rinaldo C, Tornincasa M, Gerlini R, Valente D, Izzo A, Fusco A. Hmga1 null mouse embryonic fibroblasts display downregulation of spindle assembly checkpoint gene expression associated to nuclear and karyotypic abnormalities. Cell Cycle 2016; 15:812-8. [PMID: 26889953 DOI: 10.1080/15384101.2016.1146835] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The High Mobility Group A1 proteins (HMGA1) are nonhistone chromatinic proteins with a critical role in development and cancer. We have recently reported that HMGA1 proteins are able to increase the expression of spindle assembly checkpoint (SAC) genes, thus impairing SAC function and causing chromosomal instability in cancer cells. Moreover, we found a significant correlation between HMGA1 and SAC genes expression in human colon carcinomas. Here, we report that mouse embryonic fibroblasts null for the Hmga1 gene show downregulation of Bub1, Bub1b, Mad2l1 and Ttk SAC genes, and present several features of chromosomal instability, such as nuclear abnormalities, binucleation, micronuclei and karyotypic alterations. Interestingky, also MEFs carrying only one impaired Hmga1 allele present karyotypic alterations. These results indicate that HMGA1 proteins regulate SAC genes expression and, thereby, genomic stability also in embryonic cells.
Collapse
Affiliation(s)
- Giovanna Maria Pierantoni
- a Istituto di Endocrinologia ed Oncologia Sperimentale del CNR and Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II" , Naples , Italy
| | - Andrea Conte
- a Istituto di Endocrinologia ed Oncologia Sperimentale del CNR and Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II" , Naples , Italy
| | - Cinzia Rinaldo
- b Istituto di Biologia e Patologie Molecolari del CNR c/o Università "Sapienza" di Roma , Rome , Italy
| | - Mara Tornincasa
- a Istituto di Endocrinologia ed Oncologia Sperimentale del CNR and Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II" , Naples , Italy
| | - Raffaele Gerlini
- a Istituto di Endocrinologia ed Oncologia Sperimentale del CNR and Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II" , Naples , Italy
| | - Davide Valente
- b Istituto di Biologia e Patologie Molecolari del CNR c/o Università "Sapienza" di Roma , Rome , Italy
| | - Antonella Izzo
- a Istituto di Endocrinologia ed Oncologia Sperimentale del CNR and Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II" , Naples , Italy
| | - Alfredo Fusco
- a Istituto di Endocrinologia ed Oncologia Sperimentale del CNR and Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II" , Naples , Italy
| |
Collapse
|
22
|
Conte A, Kisslinger A, Procaccini C, Paladino S, Oliviero O, de Amicis F, Faicchia D, Fasano D, Caputo M, Matarese G, Pierantoni GM, Tramontano D. Convergent Effects of Resveratrol and PYK2 on Prostate Cells. Int J Mol Sci 2016; 17:ijms17091542. [PMID: 27649143 PMCID: PMC5037816 DOI: 10.3390/ijms17091542] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/31/2016] [Accepted: 09/07/2016] [Indexed: 01/03/2023] Open
Abstract
Resveratrol, a dietary polyphenol, is under consideration as chemopreventive and chemotherapeutic agent for several diseases, including cancer. However, its mechanisms of action and its effects on non-tumor cells, fundamental to understand its real efficacy as chemopreventive agent, remain largely unknown. Proline-rich tyrosine kinase 2 (PYK2), a non-receptor tyrosine kinase acting as signaling mediator of different stimuli, behaves as tumor-suppressor in prostate. Since, PYK2 and RSV share several fields of interaction, including oxidative stress, we have investigated their functional relationship in human non-transformed prostate EPN cells and in their tumor-prone counterpart EPN-PKM, expressing a PYK2 dead-kinase mutant. We show that RSV has a strong biological activity in both cell lines, decreasing ROS production, inducing morphological changes and reversible growth arrest, and activating autophagy but not apoptosis. Interestingly, the PYK2 mutant increases basal ROS and autophagy levels, and modulates the intensity of RSV effects. In particular, the anti-oxidant effect of RSV is more potent in EPN than in EPN-PKM, whereas its anti-proliferative and pro-autophagic effects are more significant in EPN-PKM. Consistently, PYK2 depletion by RNAi replicates the effects of the PKM mutant. Taken together, our results reveal that PYK2 and RSV act on common cellular pathways and suggest that RSV effects on prostate cells may depend on mutational-state or expression levels of PYK2 that emerges as a possible mediator of RSV mechanisms of action. Moreover, the observation that resveratrol effects are reversible and not associated to apoptosis in tumor-prone EPN-PKM cells suggests caution for its use in humans.
Collapse
Affiliation(s)
- Andrea Conte
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
- Institute of Experimental Oncology and Endocrinology, National Research Council of Italy, 80131 Naples, Italy.
| | - Annamaria Kisslinger
- Institute of Experimental Oncology and Endocrinology, National Research Council of Italy, 80131 Naples, Italy.
| | - Claudio Procaccini
- Institute of Experimental Oncology and Endocrinology, National Research Council of Italy, 80131 Naples, Italy.
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
- Centro di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, 80131 Naples, Italy.
| | - Olimpia Oliviero
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, 80131 Naples, Italy.
| | - Francesca de Amicis
- Centro Sanitario, University of Calabria, 87036 Rende (CS), Italy.
- Department of Pharmacy, Health Science and Nutrition, University of Calabria, 87036 Rende (CS), Italy.
| | - Deriggio Faicchia
- Department of Medical and Translational Science, University Federico II of Naples, 80131 Naples, Italy.
| | - Dominga Fasano
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
| | - Marilena Caputo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
| | - Giuseppe Matarese
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
| | - Giovanna Maria Pierantoni
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
| | - Donatella Tramontano
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
| |
Collapse
|
23
|
Zhou WB, Zhong CN, Luo XP, Zhang YY, Zhang GY, Zhou DX, Liu LP. miR-625 suppresses cell proliferation and migration by targeting HMGA1 in breast cancer. Biochem Biophys Res Commun 2016; 470:838-44. [PMID: 26806308 DOI: 10.1016/j.bbrc.2016.01.122] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 01/20/2016] [Indexed: 12/28/2022]
Abstract
Dysregulation of microRNA contributes to the high incidence and mortality of breast cancer. Here, we show that miR-625 was frequently down-regulated in breast cancer. Decrease of miR-625 was closely associated with estrogen receptor (P = 0.004), human epidermal growth factor receptor 2 (P = 0.003) and clinical stage (P = 0.001). Kaplan-Meier and multivariate analyses indicated miR-625 as an independent factor for unfavorable prognosis (hazard ratio = 2.654, 95% confident interval: 1.300-5.382, P = 0.007). Re-expression of miR-625 impeded, whereas knockdown of miR-625 enhanced cell viabilities and migration abilities in breast cancer cells. HMGA1 was confirmed as a direct target of miR-625. The expressions of HMGA1 mRNA and protein were induced by miR-625 mimics, but reduced by miR-625 inhibitor. Re-introduction of HMGA1 in cells expressing miR-625 distinctly abrogated miR-625-mediated inhibition of cell growth. Taken together, our data demonstrate that miR-625 suppresses cell proliferation and migration by targeting HMGA1 and suggest miR-625 as a promising prognostic biomarker and a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Wen-bin Zhou
- Department of Breast Surgery, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong Province, China
| | - Cai-neng Zhong
- Department of Breast Surgery, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong Province, China
| | - Xun-peng Luo
- Department of Breast Surgery, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong Province, China
| | - Ya-yuan Zhang
- Department of Breast Surgery, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong Province, China
| | - Gui-ying Zhang
- Department of Breast Surgery, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong Province, China
| | - Dong-xian Zhou
- Department of Breast Surgery, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong Province, China.
| | - Li-ping Liu
- Department of Hepatobiliary and Pancreas Surgery, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong Province, China.
| |
Collapse
|