1
|
Petroni M, Fabretti F, Giulio SD, Robilant VND, Monica VL, Moretti M, Belardinilli F, Bufalieri F, Anna C, Paci P, Corsi A, Smaele ED, Coni S, Canettieri G, Marcotullio LD, Wang ZQ, Giannini G. A gene dosage-dependent effect unveils NBS1 as both a haploinsufficient tumour suppressor and an essential gene for SHH-medulloblastoma. Neuropathol Appl Neurobiol 2022; 48:e12837. [PMID: 35839783 PMCID: PMC9542137 DOI: 10.1111/nan.12837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/10/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Inherited or somatic mutations in the MRE11, RAD50 and NBN genes increase the incidence of tumours, including medulloblastoma (MB). On the other hand, MRE11, RAD50 and NBS1 protein components of the MRN complex are often overexpressed and sometimes essential in cancer. In order to solve the apparent conundrum about the oncosuppressive or oncopromoting role of the MRN complex, we explored the functions of NBS1 in a MB prone animal model. MATERIALS AND METHODS We generated and analysed mono- or biallelic deletion of the Nbn gene in the context of the SmoA1 transgenic mouse, a SHH-dependent MB prone animal model. We used normal and tumour tissue from these animal models, primary granule cell progenitors (GCPs) from genetically modified animals, and NBS1-depleted primary MB cells, to uncover the effects of NBS1-depletion by RNA-Seq, by biochemical characterization of the SHH-pathway and the DNA damage response (DDR) as well as on the growth and clonogenic properties of GCPs. RESULTS We found that monoallelic Nbn deletion increases SmoA1-dependent MB incidence. In addition to a defective DDR, Nbn+/- GCPs show increased clonogenicity compared to Nbn+/+ GCPs, dependent on an enhanced Notch signalling. In contrast, full NbnKO impairs MB development both in SmoA1 mice and in a SHH-driven tumour allograft. CONCLUSIONS Our study indicates that Nbn is haploinsufficient for SHH-MB development while full NbnKO is epistatic on SHH-driven MB development, thus revealing a gene dosage-dependent effect of Nbn inactivation on SHH-MB development.
Collapse
Affiliation(s)
- Marialaura Petroni
- Dept. of Molecular Medicine, University La Sapienza, Rome, Italy.,Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | | | | | | | | | - Marta Moretti
- Dept. of Experimental Medicine, University La Sapienza, Rome, Italy
| | | | | | - Coppa Anna
- Dept. of Experimental Medicine, University La Sapienza, Rome, Italy
| | - Paola Paci
- Dep. of Computer Engineering, Automation and Management, University La Sapienza, Rome, Italy.,Institute for Systems Analysis and Computer Science Antonio Ruberti, National Research Council, Rome, Italy
| | - Alessandro Corsi
- Dept. of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Enrico De Smaele
- Dept. of Experimental Medicine, University La Sapienza, Rome, Italy
| | - Sonia Coni
- Dept. of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Gianluca Canettieri
- Dept. of Molecular Medicine, University La Sapienza, Rome, Italy.,Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | | | - Zhao-Qi Wang
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI) Beutenbergstrasse 11, Jena, Germany
| | - Giuseppe Giannini
- Dept. of Molecular Medicine, University La Sapienza, Rome, Italy.,Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
2
|
Di Giulio S, Colicchia V, Pastorino F, Pedretti F, Fabretti F, Nicolis di Robilant V, Ramponi V, Scafetta G, Moretti M, Licursi V, Belardinilli F, Peruzzi G, Infante P, Goffredo BM, Coppa A, Canettieri G, Bartolazzi A, Ponzoni M, Giannini G, Petroni M. A combination of PARP and CHK1 inhibitors efficiently antagonizes MYCN-driven tumors. Oncogene 2021; 40:6143-6152. [PMID: 34508175 PMCID: PMC8553625 DOI: 10.1038/s41388-021-02003-0] [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: 09/17/2020] [Revised: 08/18/2021] [Accepted: 08/27/2021] [Indexed: 11/13/2022]
Abstract
MYCN drives aggressive behavior and refractoriness to chemotherapy, in several tumors. Since MYCN inactivation in clinical settings is not achievable, alternative vulnerabilities of MYCN-driven tumors need to be explored to identify more effective and less toxic therapies. We previously demonstrated that PARP inhibitors enhance MYCN-induced replication stress and promote mitotic catastrophe, counteracted by CHK1. Here, we showed that PARP and CHK1 inhibitors synergized to induce death in neuroblastoma cells and in primary cultures of SHH-dependent medulloblastoma, their combination being more effective in MYCN amplified and MYCN overexpressing cells compared to MYCN non-amplified cells. Although the MYCN amplified IMR-32 cell line carrying the p.Val2716Ala ATM mutation showed the highest sensitivity to the drug combination, this was not related to ATM status, as indicated by CRISPR/Cas9-based correction of the mutation. Suboptimal doses of the CHK1 inhibitor MK-8776 plus the PARP inhibitor olaparib led to a MYCN-dependent accumulation of DNA damage and cell death in vitro and significantly reduced the growth of four in vivo models of MYCN-driven tumors, without major toxicities. Our data highlight the combination of PARP and CHK1 inhibitors as a new potential chemo-free strategy to treat MYCN-driven tumors, which might be promptly translated into clinical trials.
Collapse
Affiliation(s)
- Stefano Di Giulio
- Department of Molecular Medicine, University La Sapienza, 00161, Rome, Italy
| | - Valeria Colicchia
- Department of Molecular Medicine, University La Sapienza, 00161, Rome, Italy.,Department of Biology, University Tor Vergata, 00173, Rome, Italy
| | - Fabio Pastorino
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Flaminia Pedretti
- Department of Molecular Medicine, University La Sapienza, 00161, Rome, Italy.,Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Francesca Fabretti
- Department of Molecular Medicine, University La Sapienza, 00161, Rome, Italy
| | | | - Valentina Ramponi
- Department of Molecular Medicine, University La Sapienza, 00161, Rome, Italy.,Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Giorgia Scafetta
- Pathology Research Laboratory, Sant'Andrea University Hospital, 00189, Rome, Italy
| | - Marta Moretti
- Department of Experimental Medicine, University La Sapienza, 00161, Rome, Italy
| | - Valerio Licursi
- Department of Biology and Biotechnologies "Charles Darwin", University La Sapienza, 00185, Rome, Italy
| | | | - Giovanna Peruzzi
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Paola Infante
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | | | - Anna Coppa
- Department of Experimental Medicine, University La Sapienza, 00161, Rome, Italy
| | - Gianluca Canettieri
- Department of Molecular Medicine, University La Sapienza, 00161, Rome, Italy.,Istituto Pasteur-Fondazione Cenci Bolognetti, 00161, Rome, Italy
| | - Armando Bartolazzi
- Pathology Research Laboratory, Sant'Andrea University Hospital, 00189, Rome, Italy
| | - Mirco Ponzoni
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, University La Sapienza, 00161, Rome, Italy. .,Istituto Pasteur-Fondazione Cenci Bolognetti, 00161, Rome, Italy.
| | - Marialaura Petroni
- Department of Molecular Medicine, University La Sapienza, 00161, Rome, Italy
| |
Collapse
|
3
|
Li G, Luo W, Wang B, Qian C, Ye Y, Li Y, Zhang S. HMGA1 Induction of miR-103/107 Forms a Negative Feedback Loop to Regulate Autophagy in MPTP Model of Parkinson's Disease. Front Cell Neurosci 2021; 14:620020. [PMID: 33536877 PMCID: PMC7847849 DOI: 10.3389/fncel.2020.620020] [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: 10/21/2020] [Accepted: 12/09/2020] [Indexed: 11/18/2022] Open
Abstract
Autophagy dysfunction has been directly linked with the onset and progression of Parkinson’s disease (PD), but the underlying mechanisms are not well understood. High-mobility group A1 (HMGA1), well-known chromatin remodeling proteins, play pivotal roles in diverse biological processes and diseases. Their function in neural cell death in PD, however, have not yet been fully elucidated. Here, we report that HMGA1 is highly induced during dopaminergic cell death in vitro and mice models of PD in vivo. Functional studies using genetic knockdown of endogenous HMGA1 show that HMGA1 signaling inhibition accelerates neural cell death, at least partially through aggravating MPP+-induced autophagic flux reduction resulting from partial block in autophagic flux at the terminal stages, indicating a novel potential neuroprotective role for HMGA1 in dopaminergic neurons death. MicroRNA-103/107 (miR-103/107) family, which is highly expressed in neuron, coordinately ensures proper end-stage autophagy. We further illustrate that MPP+/1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced HMGA1 elevation counterparts the effect of miR-103/107 downregulation by directly binding to their promoters, respectively, sustaining their expression in MPP+-damaged MN9D cells and modulates autophagy through CDK5R1/CDK5 signaling pathway. We also find that HMGA1 is a direct target of miR-103/107 family. Thus, our results suggest that HMGA1 forms a negative feedback loop with miR-103/107-CDK5R1/CDK5 signaling to regulate the MPP+/MPTP-induced autophagy impairment and neural cell death. Collectively, we identify a paradigm for compensatory neuroprotective HMGA1 signaling in dopaminergic neurons that could have important therapeutic implications for PD.
Collapse
Affiliation(s)
- Gehui Li
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The National Key Clinical Specialty, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Wanxian Luo
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Baoyan Wang
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The National Key Clinical Specialty, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Qian
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The National Key Clinical Specialty, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yongyi Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuantao Li
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Shizhong Zhang
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The National Key Clinical Specialty, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
De Martino M, Fusco A, Esposito F. HMGA and Cancer: A Review on Patent Literatures. Recent Pat Anticancer Drug Discov 2020; 14:258-267. [PMID: 31538905 DOI: 10.2174/1574892814666190919152001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/04/2019] [Accepted: 09/11/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND The high mobility group A proteins modulate the transcription of numerous genes by interacting with transcription factors and/or altering the structure of chromatin. These proteins are involved in both benign and malignant neoplasias as a result of several pathways. A large amount of benign human mesenchymal tumors has rearrangements of HMGA genes. On the contrary, malignant tumors show unarranged HMGA overexpression that is frequently and causally related to neoplastic cell transformation. Here, we review the function of the HMGA proteins in human neoplastic disorders, the pathways by which they contribute to carcinogenesis and the new patents focused on targeting HMGA proteins. OBJECTIVE Current review was conducted to check the involvement of HMGA as a druggable target in cancer treatment. METHODS We reviewed the most recent patents focused on targeting HMGA in cancer treatment analyzing patent literature published during the last years, including the World Intellectual Property Organization (WIPO®), United States Patent Trademark Office (USPTO®), Espacenet®, and Google Patents. RESULTS HMGA proteins are intriguing targets for cancer therapy and are objects of different patents based on the use of DNA aptamers, inhibitors, oncolytic viruses, antisense molecules able to block their oncogenic functions. CONCLUSION Powerful strategies able to selectively interfere with HMGA expression and function could represent a helpful approach in the development of new anti-cancer therapies.
Collapse
Affiliation(s)
- Marco De Martino
- Istituto di Endocrinologia e Oncologia Sperimentale-CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita degli Studi di Napoli "Federico II", via Pansini 5, Naples 80131, Italy.,Department of Psychology, University of Campania, Caserta 81100, Italy
| | - Alfredo Fusco
- Istituto di Endocrinologia e Oncologia Sperimentale-CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita degli Studi di Napoli "Federico II", via Pansini 5, Naples 80131, Italy
| | - Francesco Esposito
- Istituto di Endocrinologia e Oncologia Sperimentale-CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita degli Studi di Napoli "Federico II", via Pansini 5, Naples 80131, Italy
| |
Collapse
|
5
|
High-mobility group AT-hook 1 promotes cardiac dysfunction in diabetic cardiomyopathy via autophagy inhibition. Cell Death Dis 2020; 11:160. [PMID: 32123163 PMCID: PMC7052237 DOI: 10.1038/s41419-020-2316-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 01/09/2020] [Accepted: 01/09/2020] [Indexed: 12/12/2022]
Abstract
High-mobility group AT-hook1 (HMGA1, formerly HMG-I/Y), an architectural transcription factor, participates in a number of biological processes. However, its effect on cardiac remodeling (refer to cardiac inflammation, apoptosis and dysfunction) in diabetic cardiomyopathy remains largely indistinct. In this study, we found that HMGA1 was upregulated in diabetic mouse hearts and high-glucose-stimulated cardiomyocytes. Overexpression of HMGA1 accelerated high-glucose-induced cardiomyocyte inflammation and apoptosis, while HMGA1 knockdown relieved inflammation and apoptosis in cardiomyocytes in response to high glucose. Overexpression of HMGA1 in mice heart by adeno-associated virus 9 (AAV9) delivery system deteriorated the inflammatory response, increased apoptosis and accelerated cardiac dysfunction in streptozotocin-induced diabetic mouse model. Knockdown of HMGA1 by AAV9-shHMGA1 in vivo ameliorated cardiac remodeling in diabetic mice. Mechanistically, we found that HMGA1 inhibited the formation rather than the degradation of autophagy by regulating P27/CDK2/mTOR signaling. CDK2 knockdown or P27 overexpression blurred HMGA1 overexpression-induced deteriorating effects in vitro. P27 overexpression in mice heart counteracted HMGA1 overexpression-induced increased cardiac remodeling in diabetic mice. The luciferase reporter experiment confirmed that the regulatory effect of HMGA1 on P27 was mediated by miR-222. In addition, a miR-222 antagomir counteracted HMGA1 overexpression-induced deteriorating effects in vitro. Taken together, our data indicate that HMGA1 aggravates diabetic cardiomyopathy by directly regulating miR-222 promoter activity, which inhibits P27/mTOR-induced autophagy.
Collapse
|
6
|
Zhang W, Zhao H, Chen K, Huang Y. Overexpressing of POU2F2 accelerates fracture healing via regulating HMGA1/Wnt/β-catenin signaling pathway. Biosci Biotechnol Biochem 2019; 84:491-499. [PMID: 31782345 DOI: 10.1080/09168451.2019.1695574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
To elucidate the role of POU2F2 (POU class 2 homeobox 2) in fracture healing, 30 rats with femoral fracture were randomly grouped into three groups: FF group, LV-POU2F2 group and LV-scramble group. Rats were injected with PBS, lentivirus expressing POU2F2 or scramble lentivirus once a week for 4 weeks. Results showed that overexpressing of POU2F2 promoted fracture healing and callus growth. Besides, overexpressing of POU2F2 promoted protein and mRNA expression of Col10a1, Runx2, Osterix, and Osteocalcin. High Mobility Group AT-hook 1 (HMGA1) is a non-histone protein participating in chromatin remodeling of cells. Western blotting manifested HMGA1/Wnt/β-catenin pathway was activated in POU2F2 group. Moreover, in-vitro study of hMSCs cells supported the above data. In conclusion, POU2F2 promotes fracture healing via activating the HMGA1/Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hanke Zhao
- Department of Orthopedics, Changzhou Wujin People's Hospital, Changzhou, China
| | - Kun Chen
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ye Huang
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Lindholm EM, Ragle Aure M, Haugen MH, Kleivi Sahlberg K, Kristensen VN, Nebdal D, Børresen‐Dale A, Lingjærde OC, Engebraaten O. miRNA expression changes during the course of neoadjuvant bevacizumab and chemotherapy treatment in breast cancer. Mol Oncol 2019; 13:2278-2296. [PMID: 31402562 PMCID: PMC6763780 DOI: 10.1002/1878-0261.12561] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/26/2019] [Accepted: 08/09/2019] [Indexed: 12/24/2022] Open
Abstract
One of the hallmarks of cancer is sustained angiogenesis. Favorable results have been reported in some breast cancer (BC) patients receiving antiangiogenic therapy with bevacizumab (Bev) in combination with chemotherapy, and further knowledge on how Bev can be optimally combined with conventional treatment to increase efficacy is strongly needed. In this randomized, neoadjuvant phase II clinical trial, 132 patients with HER2-negative, nonmetastatic BC were treated with Bev in combination with sequential chemotherapy. Biopsies were sampled before treatment, after 12 weeks with anthracycline and after taxane therapy at week 25. MicroRNA (miRNA) expression profiling was performed on biopsies from each time point. Altogether, 241 biopsies were analyzed with the aim of identifying miRNA-based biomarkers of response to therapy. Results from the miRNA analyses were reported for the ER-positive cohort, which were previously demonstrated to benefit from antiangiogenic therapy in this study. For both treatment arms of this cohort, significantly different expression was observed for 217 miRNAs between objective responding and nonresponding patients before treatment initiation. These miRNAs have been linked to regulation of epithelial-mesenchymal transition, metastasis, and tumor growth, among other processes. Bev in combination with chemotherapy resulted in similar miRNA changes to chemotherapy alone. However, the deregulation of miRNA expression occurred earlier in the Bev arm. In both arms, tumor suppressor miRNAs were found upregulated after treatment, while oncogenic miRNAs were downregulated in the Bev arm. Patients responding to Bev showed a strong correlation between deregulated miRNAs and decreased proliferation score during the course of treatment, with downregulation of miR-4465 as the strongest indicator of reduced proliferation. Integrative analyses at miRNA-, gene-, and protein expression further indicated a longitudinal decrease in proliferation. Altogether, the results indicate that proliferation might represent a predictive factor for increased Bev sensitivity, which may aid in the identification of patients who could potentially benefit from Bev.
Collapse
Affiliation(s)
- Evita Maria Lindholm
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
| | - Miriam Ragle Aure
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
| | - Mads Haugland Haugen
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Department of Tumor biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
| | - Kristine Kleivi Sahlberg
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Department of Research and InnovationVestre Viken Hospital TrustDrammenNorway
| | - Vessela N. Kristensen
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Department of Clinical Molecular Biology (EpiGen), Division of MedicineAkershus University HospitalLørenskogNorway
| | - Daniel Nebdal
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
| | - Anne‐Lise Børresen‐Dale
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Insitute for Clinical MedicineUniversity of OsloNorway
| | - Ole Christian Lingjærde
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Department of Computer Science, Faculty of Mathematics and Natural SciencesUniversity of OsloNorway
| | - Olav Engebraaten
- Department of Tumor biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalNorway
- Insitute for Clinical MedicineUniversity of OsloNorway
- Department of OncologyOslo University HospitalNorway
| |
Collapse
|
8
|
Fu F, Wang T, Wu Z, Feng Y, Wang W, Zhou S, Ma X, Wang S. HMGA1 exacerbates tumor growth through regulating the cell cycle and accelerates migration/invasion via targeting miR-221/222 in cervical cancer. Cell Death Dis 2018; 9:594. [PMID: 29789601 PMCID: PMC5964147 DOI: 10.1038/s41419-018-0683-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/11/2018] [Accepted: 05/03/2018] [Indexed: 12/17/2022]
Abstract
High-mobility group AT-hook1 (HMGA1, formerly HMG-I/Y), an architectural transcription factor, participates in a number of tumor biological processes. However, its effect on cervical cancer remains largely indistinct. In this study, we found that HMGA1 was generally overexpressed in cervical cancer tissues and was positively correlated with lymph node metastasis and advanced clinical stage. Via exogenously increasing or decreasing the expression of HMGA1, we showed that HMGA1 affected the proliferation, colony formation, migration and invasion of cervical cancer cells in vitro. Rescue experiments suggested that miR-221/222 could partly reverse HMGA1-mediated migration and invasion processes. Mechanistically, we discovered that HMGA1 accelerated the G1/S phase transition by regulating the expression of cyclin D1 and cyclin E1, which was consistent with the results of the in vivo experiment. Furthermore, we found that HMGA1 regulated the expression of the miR-221/222 cluster at the transcriptional level and that miR-221/222 targeted the 3'UTR of tissue inhibitor of metalloproteinases 3(TIMP3). We propose a fresh perspective that HMGA1 participates in the migration and invasion process via the miR-221/222-TIMP3-MMP2/MMP9 axis in cervical cancer. In summary, our study identified a critical role played by HMGA1 in the progression of cervical cancer and the potential mechanisms by which exerts its effects, suggesting that targeting HMGA1-related pathways could be conducive to the therapies for cervical cancer.
Collapse
Affiliation(s)
- Fangfang Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Tian Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Zhangying Wu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, 55000, Guiyang, Guizhou, China
| | - Yourong Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Wenwen Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Su Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Xiangyi Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China.
| |
Collapse
|
9
|
Suman S, Mishra A, Kulshrestha A. A systems approach for the elucidation of crucial genes and network constituents of cervical intraepithelial neoplasia 1 (CIN1). MOLECULAR BIOSYSTEMS 2017; 13:549-555. [DOI: 10.1039/c6mb00615a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Network based approach for finding biomarkers for predicting the clinical behavior of cervical neoplasia cells at its earliest onset.
Collapse
Affiliation(s)
- Shikha Suman
- Division of Applied Sciences
- Indian Institute of Information Technology (IIIT)
- Allahabad-211012
- India
| | - Ashutosh Mishra
- Division of Applied Sciences
- Indian Institute of Information Technology (IIIT)
- Allahabad-211012
- India
| | | |
Collapse
|
10
|
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
|
11
|
Akhter MZ, Rajeswari MR. Interaction of doxorubicin with a regulatory element of hmga1 and its in vitro anti-cancer activity associated with decreased HMGA1 expression. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2014; 141:36-46. [DOI: 10.1016/j.jphotobiol.2014.08.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/20/2014] [Accepted: 08/23/2014] [Indexed: 11/26/2022]
|
12
|
Petkova R, Tummala H, Zhelev N. Nothing in Excess—Lessons Learned from the Expression of High-Mobility Group Proteins Type a in Non-Cancer and Cancer Cells. BIOTECHNOL BIOTEC EQ 2014. [DOI: 10.5504/bbeq.2011.0102] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
13
|
Huso TH, Resar LMS. The high mobility group A1 molecular switch: turning on cancer - can we turn it off? Expert Opin Ther Targets 2014; 18:541-53. [PMID: 24684280 DOI: 10.1517/14728222.2014.900045] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Emerging evidence demonstrates that the high mobility group A1 (HMGA1) chromatin remodeling protein is a key molecular switch required by cancer cells for tumor progression and a poorly differentiated, stem-like state. Because the HMGA1 gene and proteins are expressed at high levels in all aggressive tumors studied to date, research is needed to determine how to 'turn off' this master regulatory switch in cancer. AREAS COVERED In this review, we describe prior studies that underscore the central role of HMGA1 in refractory cancers and we discuss approaches to target HMGA1 in cancer therapy. EXPERT OPINION Given the widespread overexpression of HMGA1 in diverse, aggressive tumors, further research to develop technology to target HMGA1 holds immense promise as potent anticancer therapy. Previous work in preclinical models indicates that delivery of short hairpin RNA or interfering RNA molecules to 'switch off' HMGA1 expression dramatically impairs cancer cell growth and tumor progression. The advent of nanoparticle technology to systemically deliver DNA or RNA molecules to tumors brings this approach even closer to clinical applications, although further efforts are needed to translate these advances into therapies for cancer patients.
Collapse
Affiliation(s)
- Tait H Huso
- The Johns Hopkins University School of Medicine, Hematology Division , Ross Research Building, Room 1015, 720 Rutland Avenue, Baltimore MD 21205 , USA
| | | |
Collapse
|
14
|
Veschi V, Petroni M, Bartolazzi A, Altavista P, Dominici C, Capalbo C, Boldrini R, Castellano A, McDowell HP, Pizer B, Frati L, Screpanti I, Gulino A, Giannini G. Galectin-3 is a marker of favorable prognosis and a biologically relevant molecule in neuroblastic tumors. Cell Death Dis 2014; 5:e1100. [PMID: 24603328 PMCID: PMC3973198 DOI: 10.1038/cddis.2014.68] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/16/2014] [Accepted: 01/22/2014] [Indexed: 01/03/2023]
Abstract
Childhood neuroblastic tumors are characterized by heterogeneous clinical courses, ranging from benign ganglioneuroma (GN) to highly lethal neuroblastoma (NB). Although a refined prognostic evaluation and risk stratification of each tumor patient is becoming increasingly essential to personalize treatment options, currently only few biomolecular markers (essentially MYCN amplification, chromosome 11q status and DNA ploidy) are validated for this purpose in neuroblastic tumors. Here we report that Galectin-3 (Gal-3), a β-galactoside-binding lectin involved in multiple biological functions that has already acquired diagnostic relevance in specific clinical settings, is variably expressed in most differentiated and less aggressive neuroblastic tumors, such as GN and ganglioneuroblastoma, as well as in a subset of NB cases. Gal-3 expression is associated with the INPC histopathological categorization (P<0.001) and Shimada favorable phenotype (P=0.001), but not with other prognostically relevant features. Importantly, Gal-3 expression was associated with a better 5-year overall survival (P=0.003), and with improved cumulative survival in patient subsets at worse prognosis, such as older age at diagnosis, advanced stages or NB histopathological classification. In vitro, Gal-3 expression and nuclear accumulation accompanied retinoic acid-induced cell differentiation in NB cell lines. Forced Gal-3 overexpression increased phenotypic differentiation and substrate adherence, while inhibiting proliferation. Altogether, these findings suggest that Gal-3 is a biologically relevant player for neuroblastic tumors, whose determination by conventional immunohistochemistry might be used for outcome assessment and patient's risk stratification in the clinical setting.
Collapse
Affiliation(s)
- V Veschi
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - M Petroni
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - A Bartolazzi
- 1] Department of Pathology, St. Andrea Hospital, Rome, Italy [2] Pathology Research Laboratory, Cancer Center Karolinska (CCK), Karolinska Hospital, Stockholm, Sweden
| | - P Altavista
- Unit of Radiation Biology and Human Health, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Research Center Casaccia, Rome, Italy
| | - C Dominici
- 1] Department of Pediatrics and Infantile Neuropsychiatry, University La Sapienza, Rome, Italy [2] School of Reproductive and Developmental Medicine, Liverpool University, Liverpool, UK
| | - C Capalbo
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - R Boldrini
- Division of Pathology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - A Castellano
- Division of Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - H P McDowell
- 1] Department of Pediatrics and Infantile Neuropsychiatry, University La Sapienza, Rome, Italy [2] School of Reproductive and Developmental Medicine, Liverpool University, Liverpool, UK
| | - B Pizer
- Department of Oncology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - L Frati
- Department of Experimental Medicine, University La Sapienza, Rome, Italy
| | - I Screpanti
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - A Gulino
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - G Giannini
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| |
Collapse
|
15
|
Sompallae R, Hofmann O, Maher CA, Gedye C, Behren A, Vitezic M, Daub CO, Devalle S, Caballero OL, Carninci P, Hayashizaki Y, Lawlor ER, Cebon J, Hide W. A comprehensive promoter landscape identifies a novel promoter for CD133 in restricted tissues, cancers, and stem cells. Front Genet 2013; 4:209. [PMID: 24194746 PMCID: PMC3810939 DOI: 10.3389/fgene.2013.00209] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 09/30/2013] [Indexed: 12/02/2022] Open
Abstract
PROM1 is the gene encoding prominin-1 or CD133, an important cell surface marker for the isolation of both normal and cancer stem cells. PROM1 transcripts initiate at a range of transcription start sites (TSS) associated with distinct tissue and cancer expression profiles. Using high resolution Cap Analysis of Gene Expression (CAGE) sequencing we characterize TSS utilization across a broad range of normal and developmental tissues. We identify a novel proximal promoter (P6) within CD133+ melanoma cell lines and stem cells. Additional exon array sampling finds P6 to be active in populations enriched for mesenchyme, neural stem cells and within CD133+ enriched Ewing sarcomas. The P6 promoter is enriched with respect to previously characterized PROM1 promoters for a HMGI/Y (HMGA1) family transcription factor binding site motif and exhibits different epigenetic modifications relative to the canonical promoter region of PROM1.
Collapse
|
16
|
Veschi V, Petroni M, Cardinali B, Dominici C, Screpanti I, Frati L, Bartolazzi A, Gulino A, Giannini G. Galectin-3 impairment of MYCN-dependent apoptosis-sensitive phenotype is antagonized by nutlin-3 in neuroblastoma cells. PLoS One 2012; 7:e49139. [PMID: 23152863 PMCID: PMC3494673 DOI: 10.1371/journal.pone.0049139] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 10/03/2012] [Indexed: 11/18/2022] Open
Abstract
MYCN amplification occurs in about 20–25% of human neuroblastomas and characterizes the majority of the high-risk cases, which display less than 50% prolonged survival rate despite intense multimodal treatment. Somehow paradoxically, MYCN also sensitizes neuroblastoma cells to apoptosis, understanding the molecular mechanisms of which might be relevant for the therapy of MYCN amplified neuroblastoma. We recently reported that the apoptosis-sensitive phenotype induced by MYCN is linked to stabilization of p53 and its proapoptotic kinase HIPK2. In MYCN primed neuroblastoma cells, further activation of both HIPK2 and p53 by Nutlin-3 leads to massive apoptosis in vitro and to tumor shrinkage and impairment of metastasis in xenograft models. Here we report that Galectin-3 impairs MYCN-primed and HIPK2-p53-dependent apoptosis in neuroblastoma cells. Galectin-3 is broadly expressed in human neuroblastoma cell lines and tumors and is repressed by MYCN to induce the apoptosis-sensitive phenotype. Despite its reduced levels, Galectin-3 can still exert residual antiapoptotic effects in MYCN amplified neuroblastoma cells, possibly due to its specific subcellular localization. Importantly, Nutlin-3 represses Galectin-3 expression, and this is required for its potent cell killing effect on MYCN amplified cell lines. Our data further characterize the apoptosis-sensitive phenotype induced by MYCN, expand our understanding of the activity of MDM2-p53 antagonists and highlight Galectin-3 as a potential biomarker for the tailored p53 reactivation therapy in patients with high-risk neuroblastomas.
Collapse
Affiliation(s)
- Veronica Veschi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | - Beatrice Cardinali
- Institute of Cell Biology and Neurobiology, National Research Council, Monterotondo Scalo, Italy
| | - Carlo Dominici
- Department of Pediatrics, Sapienza University, Rome, Italy
- School of Reproductive and Developmental Medicine, Liverpool University, Liverpool, United Kingdom
| | | | - Luigi Frati
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Armando Bartolazzi
- Experimental Pathology Laboratory, S. Andrea Hospital, Rome, Italy
- Cancer Center Karolinska (CCK) R8∶04, Karolinska Hospital, Stockholm, Sweden
| | - Alberto Gulino
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, Sapienza University, Rome, Italy
- * E-mail:
| |
Collapse
|
17
|
Massimi I, Guerrieri F, Petroni M, Veschi V, Truffa S, Screpanti I, Frati L, Levrero M, Gulino A, Giannini G. The HMGA1 protoncogene frequently deregulated in cancer is a transcriptional target of E2F1. Mol Carcinog 2012; 52:526-34. [PMID: 22389255 DOI: 10.1002/mc.21887] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 12/05/2011] [Accepted: 01/25/2012] [Indexed: 01/28/2023]
Abstract
Reactivation of the HMGA1 protoncogene is very frequent in human cancer, but still very little is known on the molecular mechanisms leading to this event. Prompted by the finding of putative E2F binding sites in the human HMGA1 promoter and by the frequent deregulation of the RB/E2F1 pathway in human carcinogenesis, we investigated whether E2F1 might contribute to the regulation of HMGA1 gene expression. Here we report that E2F1 induces HMGA1 by interacting with a 193 bp region of the HMGA1 promoter containing an E2F binding site surrounded by three putative Sp1 binding sites. Both gain and loss of function experiments indicate that Sp1 functionally interacts with E2F1 to promote HMGA1 expression. However, while Sp1 constitutively binds HMGA1 promoter, it is the balance between different E2F family members that tunes the levels of HMGA1 expression between quiescence and proliferation. Finally, we found increased HMGA1 expression in pituitary and thyroid tumors developed in Rb(+/-) mice, supporting the hypothesis that E2F1 is a novel important regulator of HMGA1 expression and that deregulation of the RB/E2F1 path might significantly contribute to HMGA1 deregulation in cancer.
Collapse
Affiliation(s)
- Isabella Massimi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Barh D, Malhotra R, Ravi B, Sindhurani P. MicroRNA let-7: an emerging next-generation cancer therapeutic. ACTA ACUST UNITED AC 2011; 17:70-80. [PMID: 20179807 PMCID: PMC2826782 DOI: 10.3747/co.v17i1.356] [Citation(s) in RCA: 203] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, various RNA-based technologies have been under evaluation as potential next-generation cancer therapeutics. Micrornas (miRNAS), known to regulate the cell cycle and development, are deregulated in various cancers. Thus, they might serve as good targets or candidates in an exploration of anticancer therapeutics. One attractive candidate for this purpose is let-7 ("lethal-7"). Let-7 is underexpressed in various cancers, and restoration of its normal expression is found to inhibit cancer growth by targeting various oncogenes and inhibiting key regulators of several mitogenic pathways. In vivo, let-7 administration was found effective against mouse-model lung and breast cancers, and our computational prediction supports the possible effectiveness of let-7 in estrogen receptor (ER)-positive metastatic breast cancer. Data also suggest that let-7 regulates apoptosis and cancer stem cell (CSC) differentiation and can therefore be tested as a potential therapeutic in cancer treatment. However, the exact role of let-7 in cancer is not yet fully understood. There is a need to understand the causative molecular basis of let-7 alterations in cancer and to develop proper delivery systems before proceeding to therapeutic applications. This article attempts to highlight certain critical aspects of let-7's therapeutic potential in cancer.
Collapse
Affiliation(s)
- D Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, India.
| | | | | | | |
Collapse
|
19
|
Differential expression and prognostic value of HMGA1 in pancreatic head and periampullary cancer. Eur J Cancer 2010; 46:3393-9. [DOI: 10.1016/j.ejca.2010.07.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 07/15/2010] [Indexed: 11/22/2022]
|
20
|
High mobility group A: A novel biomarker and therapeutic target in pancreatic adenocarcinoma. Surgeon 2009; 7:297-306. [DOI: 10.1016/s1479-666x(09)80008-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
21
|
De Martino I, Visone R, Fedele M, Petrocca F, Palmieri D, Martinez Hoyos J, Forzati F, Croce CM, Fusco A. Regulation of microRNA expression by HMGA1 proteins. Oncogene 2009; 28:1432-42. [PMID: 19169275 DOI: 10.1038/onc.2008.495] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The High Mobility Group proteins HMGA1 are nuclear architectural factors that play a critical role in a wide range of biological processes. Since recent studies have identified the microRNAs (miRNAs) as important regulators of gene expression, modulating critical cellular functions such as proliferation, apoptosis and differentiation, the aim of our work was to identify the miRNAs that are physiologically regulated by HMGA1 proteins. To this purpose, we have analysed the miRNA expression profile of mouse embryonic fibroblasts (MEFs) carrying two, one or no Hmga1 functional alleles using a microarray (miRNA microarray). By this approach, we found a miRNA expression profile that differentiates Hmga1-null MEFs from the wild-type ones. In particular, a significant decrease in miR-196a-2, miR-101b, miR-331 and miR-29a was detected in homozygous Hmga1-knockout MEFs in comparison with wild-type cells. Consistently, these miRNAs are downregulated in most of the analysed tissues of Hmga1-null mice in comparison with the wild-type mice. ChIP assay shows that HMGA1 is able to bind regions upstream of these miRNAs. Moreover, we identified the HMGA2 gene product as a putative target of miR-196a-2, suggesting that HMGA1 proteins are able to downregulate the expression of the other member of the HMGA family through the regulation of the miR-196a-2 expression. Finally, ATXN1 and STC1 gene products have been identified as targets of miR-101b. Therefore, it is reasonable to hypothesize that HMGA1 proteins are involved in several functions by regulating miRNA expression.
Collapse
Affiliation(s)
- I De Martino
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR c/o Dipartimento di Biologia e Patologia Cellulare e Molecolare, Facoltà di Medicina e Chirurgia di Napoli, Università degli Studi di Napoli Federico II, Naples, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Schickel R, Boyerinas B, Park SM, Peter ME. MicroRNAs: key players in the immune system, differentiation, tumorigenesis and cell death. Oncogene 2008; 27:5959-74. [PMID: 18836476 DOI: 10.1038/onc.2008.274] [Citation(s) in RCA: 593] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Micro (mi)RNAs are small, highly conserved noncoding RNAs that control gene expression post-transcriptionally either via the degradation of target mRNAs or the inhibition of protein translation. Each miRNA is believed to regulate the expression of multiple mRNA targets, and many miRNAs have been linked to the initiation and progression of human cancer. miRNAs control various activities of the immune system and different stages of hematopoietic development, and their misexpression is the cause of various blood malignancies. Certain miRNAs have oncogenic activities, whereas others have the potential to act as tumor suppressors. Because they control fundamental processes such as differentiation, cell growth and cell death, the study of the role of miRNAs in human neoplasms holds great promise for novel forms of therapy. Here, we summarize the role of miRNAs and their targets in contributing to human cancers and their function as regulators of apoptotic pathways and the immune system.
Collapse
Affiliation(s)
- R Schickel
- The Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
23
|
Human papilloma virus-dependent HMGA1 expression is a relevant step in cervical carcinogenesis. Neoplasia 2008; 10:773-81. [PMID: 18670638 DOI: 10.1593/neo.08462] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 05/12/2008] [Accepted: 05/13/2008] [Indexed: 11/18/2022] Open
Abstract
HMGA1 is a member of a small family of architectural transcription factors involved in the coordinate assembly of multiprotein complexes referred to as enhanceosomes. In addition to their role in cell proliferation, differentiation, and development, high-mobility group proteins of the A type (HMGA) family members behave as transforming protoncogenes either in vitro or in animal models. Recent reports indicated that HMGA1 might counteract p53 pathway and provided an interesting hint on the mechanisms determining HMGA's transforming potential. HMGA1 expression is deregulated in a very large array of human tumors, including cervical cancer, but very limited information is available on the molecular mechanisms leading to HMGA1 deregulation in cancer cells. Here, we report that HMGA1 expression is sustained by human papilloma virus (HPV) E6/E7 proteins in cervical cancer, as demonstrated by either E6/E7 overexpression or by repression through RNA interference. Knocking down HMGA1 expression by means of RNA interference, we also showed that it is involved in cell proliferation and contributes to p53 inactivation in this type of neoplasia. Finally, we show that HMGA1 is necessary for the full expression of HPV18 E6 and E7 oncoproteins thus establishing a positive autoregulatory loop between HPV E6/E7 and HMGA1 expression.
Collapse
|
24
|
Abstract
Death receptors induce apoptosis through either the Type I or II pathway. In Type I cells, the initiator caspase-8 directly activates effector caspases such as caspase-3, whereas in Type II cells, the death signal is amplified through mitochondria thereby activating effector caspases causing cell death. Recently, there have been advances in elucidating the early events in the CD95 signaling pathways and how post-translational modifications regulate CD95 signaling. This review will focus on recent insights into the mechanisms of the two different types of CD95 signaling pathways, and will introduce miRNAs as regulators of death receptor signaling.
Collapse
Affiliation(s)
- Sun-Mi Park
- The Ben May Department for Cancer Research, The University of Chicago, 924 E 57th Street, Chicago, IL 60637, Phone: 773-702-4728, FAX: 773-702-3701
| | - Marcus E. Peter
- The Ben May Department for Cancer Research, The University of Chicago, 924 E 57th Street, Chicago, IL 60637, Phone: 773-702-4728, FAX: 773-702-3701
| |
Collapse
|
25
|
Koppen A, Ait-Aissa R, Koster J, Øra I, Bras J, van Sluis PG, Caron H, Versteeg R, Valentijn LJ. Dickkopf-3 expression is a marker for neuroblastic tumor maturation and is down-regulated by MYCN. Int J Cancer 2007; 122:1455-64. [DOI: 10.1002/ijc.23180] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
26
|
Smith JC, Duchesne MA, Tozzi P, Ethier M, Figeys D. A Differential Phosphoproteomic Analysis of Retinoic Acid-Treated P19 Cells. J Proteome Res 2007; 6:3174-86. [PMID: 17622165 DOI: 10.1021/pr070122r] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
External stimuli trigger internal signaling events within a cell that may represent either a temporary or permanent shift in the phosphorylation state of its proteome. Numerous reports have elucidated phosphorylation sites from a variety of biological samples and more recent studies have monitored the temporal dynamics of protein phosphorylation as a given system is perturbed. Understanding which proteins are phosphorylated as well as when they are phosphorylated may indicate novel functional roles within a system and allow new therapeutic avenues to be explored. To elucidate the dynamics of protein phosphorylation within differentiating murine P19 embryonal carcinoma cells, we induced P19 cells to differentiate using all-trans-retinoic acid and developed a strategy that combines isotopically labeled methyl esterification, immobilized metal affinity chromatography, mass spectrometric analysis, and a rigorous and unique data evaluation approach. We present the largest differential phosphoproteomic analysis using isotopically labeled methyl esterification to date, identifying a total of 472 phosphorylation sites on 151 proteins; 56 of these proteins had altered abundances following treatment with retinoic acid and approximately one-third of these have been previously associated with cellular differentiation. A series of bioinformatic tools were used to extract information from the data and explore the implications of our findings. This study represents the first global gel-free analysis that elucidates protein phosphorylation dynamics during cellular differentiation.
Collapse
Affiliation(s)
- Jeffrey C Smith
- Ottawa Institute of Systems Biology and Biochemistry, Microbiology and Immunology Department, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | | | | | | | | |
Collapse
|
27
|
Shell S, Park SM, Radjabi AR, Schickel R, Kistner EO, Jewell DA, Feig C, Lengyel E, Peter ME. Let-7 expression defines two differentiation stages of cancer. Proc Natl Acad Sci U S A 2007; 104:11400-5. [PMID: 17600087 PMCID: PMC2040910 DOI: 10.1073/pnas.0704372104] [Citation(s) in RCA: 356] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The early phases of carcinogenesis resemble embryonic development, often involving the reexpression of embryonic mesenchymal genes. The NCI60 panel of human tumor cell lines can genetically be subdivided into two superclusters (SCs) that correspond to CD95 Type I and II cells. SC1 cells are characterized by a mesenchymal and SC2 cells by an epithelial gene signature, suggesting that SC1 cells represent less differentiated, advanced stages of cancer. miRNAs are small 20- to 22-nucleotide-long noncoding RNAs that inhibit gene expression at the posttranscriptional level. By performing miRNA expression analysis on 10 Type I and 10 Type II cells, we have determined that SC1 cells express low and SC2 cells high levels of the miRNA let-7, respectively, suggesting that let-7 is a marker for less advanced cancers. Expression of the let-7 target high-mobility group A2 (HMGA2), an early embryonic gene, but not of classical epithelial or mesenchymal markers such as E-cadherin or vimentin, inversely correlated with let-7 expression in SC1 and SC2 cells. Using ovarian cancer as a model, we demonstrate that expression of let-7 and HMGA2 is a better predictor of prognosis than classical markers such as E-cadherin, vimentin, and Snail. These data identify loss of let-7 expression as a marker for less differentiated cancer.
Collapse
Affiliation(s)
| | | | - Amir Reza Radjabi
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology
| | | | - Emily O. Kistner
- Biostatistics Consulting Laboratory, Department of Health Studies, University of Chicago, Chicago, IL 60637; and
| | - David A. Jewell
- Department of Genetics, Dartmouth Medical School, Lebanon, NH 03755
| | | | - Ernst Lengyel
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology
| | - Marcus E. Peter
- *Ben May Department for Cancer Research
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
28
|
Wei T, Geiser AG, Qian HR, Su C, Helvering LM, Kulkarini NH, Shou J, N'Cho M, Bryant HU, Onyia JE. DNA microarray data integration by ortholog gene analysis reveals potential molecular mechanisms of estrogen-dependent growth of human uterine fibroids. BMC WOMENS HEALTH 2007; 7:5. [PMID: 17407572 PMCID: PMC1852551 DOI: 10.1186/1472-6874-7-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2006] [Accepted: 04/02/2007] [Indexed: 01/28/2023]
Abstract
Background Uterine fibroids or leiomyoma are a common benign smooth muscle tumor. The tumor growth is well known to be estrogen-dependent. However, the molecular mechanisms of its estrogen-dependency is not well understood. Methods Differentially expressed genes in human uterine fibroids were either retrieved from published papers or from our own statistical analysis of downloaded array data. Probes for the same genes on different Affymetrix chips were mapped based on probe comparison information provided by Affymetrix. Genes identified by two or three array studies were submitted for ortholog analysis. Human and rat ortholog genes were identified by using ortholog gene databases, HomoloGene and TOGA and were confirmed by synteny analysis with MultiContigView tool in the Ensembl genome browser. Results By integrated analysis of three recently published DNA microarray studies with human tissue, thirty-eight genes were found to be differentially expressed in the same direction in fibroid compared to adjacent uterine myometrium by at least two research groups. Among these genes, twelve with rat orthologs were identified as estrogen-regulated from our array study investigating uterine expression in ovariectomized rats treated with estrogen. Functional and pathway analyses of the twelve genes suggested multiple molecular mechanisms for estrogen-dependent cell survival and tumor growth. Firstly, estrogen increased expression of the anti-apoptotic PCP4 gene and suppressed the expression of growth inhibitory receptors PTGER3 and TGFBR2. Secondly, estrogen may antagonize PPARγ signaling, thought to inhibit fibroid growth and survival, at two points in the PPAR pathway: 1) through increased ANXA1 gene expression which can inhibit phospholipase A2 activity and in turn decrease arachidonic acid synthesis, and 2) by decreasing L-PGDS expression which would reduce synthesis of PGJ2, an endogenous ligand for PPARγ. Lastly, estrogen affects retinoic acid (RA) synthesis and mobilization by regulating expression of CRABP2 and ALDH1A1. RA has been shown to play a significant role in the development of uterine fibroids in an animal model. Conclusion Integrated analysis of multiple array datasets revealed twelve human and rat ortholog genes that were differentially expressed in human uterine fibroids and transcriptionally responsive to estrogen in the rat uterus. Functional and pathway analysis of these genes suggest multiple potential molecular mechanisms for the poorly understood estrogen-dependent growth of uterine fibroids. Fully understanding the exact molecular interactions among these gene products requires further study to validate their roles in uterine fibroids. This work provides new avenues of study which could influence the future direction of therapeutic intervention for the disease.
Collapse
Affiliation(s)
- Tao Wei
- Integrative Biology, Lilly Research Laboratories, Greenfield, Indiana 46140, USA
| | - Andrew G Geiser
- Bone and Inflammation, Lilly Research Laboratories, Indianapolis, Indiana 46285, USA
| | - Hui-Rong Qian
- Discovery Statistics, Lilly Research Laboratories, Indianapolis, Indiana 46285, USA
| | - Chen Su
- Integrative Biology, Lilly Research Laboratories, Greenfield, Indiana 46140, USA
| | - Leah M Helvering
- Bone and Inflammation, Lilly Research Laboratories, Indianapolis, Indiana 46285, USA
| | - Nalini H Kulkarini
- Integrative Biology, Lilly Research Laboratories, Greenfield, Indiana 46140, USA
| | - Jianyong Shou
- Integrative Biology, Lilly Research Laboratories, Greenfield, Indiana 46140, USA
| | - Mathias N'Cho
- Integrative Biology, Lilly Research Laboratories, Greenfield, Indiana 46140, USA
| | - Henry U Bryant
- Bone and Inflammation, Lilly Research Laboratories, Indianapolis, Indiana 46285, USA
| | - Jude E Onyia
- Integrative Biology, Lilly Research Laboratories, Greenfield, Indiana 46140, USA
| |
Collapse
|
29
|
Giannini G, Cerignoli F, Mellone M, Massimi I, Ambrosi C, Rinaldi C, Gulino A. Molecular mechanism of HMGA1 deregulation in human neuroblastoma. Cancer Lett 2005; 228:97-104. [PMID: 15923078 DOI: 10.1016/j.canlet.2005.01.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2004] [Accepted: 01/12/2005] [Indexed: 11/21/2022]
Abstract
Very soon after their original identification in HeLa cells in 1983, HMGA proteins appeared as interesting cancer-related molecules. Indeed, they were immediately noted as a sub-class of High Mobility Group proteins induced in fibroblast or epithelial cells transformed with sarcoma viruses. After more than 20 years, the association between HMGA protein expressions and cellular transformation has been largely confirmed and HMGA are among the most widely expressed cancer-associated proteins. Nevertheless, their functional contribution to tumour development and progression is far from being completely understood. Furthermore, although HMGA1 expression has been reported to be inducible by a number of factors and circumstances, the question of how their expression is deregulated in cancer is even less clear and somehow has been ignored from most researchers. An active AP1 site is the only characterized element of the HMGA1 human promoter, that remains a rather complicated and unexplored source of information to answer this question. Following the indication that c-Myc might bind and activate the mouse HMGA1 gene promoter, we have demonstrated that HMGA1 is a new target for MYCN in human neuroblastomas. In this report, we overview part of the current information on HMGA1 and focus our attention on the analysis of its human promoter.
Collapse
Affiliation(s)
- Giuseppe Giannini
- Department of Experimental Medicine and Pathology, Department of Pediatrics, University La Sapienza, Policlinico Umberto 1, Viale Regina Elena, 324, 00161 Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
30
|
Giannini G, Cerignoli F, Mellone M, Massimi I, Ambrosi C, Rinaldi C, Dominici C, Frati L, Screpanti I, Gulino A. High Mobility Group A1 Is a Molecular Target for MYCN in Human Neuroblastoma. Cancer Res 2005; 65:8308-16. [PMID: 16166307 DOI: 10.1158/0008-5472.can-05-0607] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
High mobility group A1 (HMGA1) is an architectural transcription factor and a putative protoncogene. Deregulation of its expression has been shown in most human cancers. We have previously shown that the expression of the HMGA family members is deregulated in neuroblastoma cell lines and primary tumors. On retinoic acid (RA) treatment of MYCN-amplified neuroblastoma cell lines, HMGA1 decreases with a kinetics that strictly follows MYCN repression. In addition, MYCN constitutive expression abolishes HMGA1 repression by RA. Here we explored the possibility that HMGA1 expression might be sustained by MYCN in amplified cells. Indeed, MYCN transfection induced HMGA1 expression in several neuroblastoma cell lines. HMGA1 expression increased in a transgene dose-dependent fashion in neuroblastoma-like tumors of MYCN transgenic mice. In addition, it was significantly more expressed in MYCN-amplified compared with MYCN single-copy primary human neuroblastomas. MYCN cotransfection activated a promoter/luciferase reporter containing a 1,600 bp region surrounding the first three transcription start sites of the human HMGA1 and eight imperfect E-boxes. By heterodimerizing with its partner MAX, MYCN could bind to multiple DNA fragments within the 1,600 bp. Either 5' or 3' deletion variants of the 1,600 bp promoter/luciferase reporter strongly decreased luciferase activity, suggesting that, more than a single site, the cooperative function of multiple cis-acting elements mediates direct HMGA1 transactivation by MYCN. Finally, HMGA1 repression by RNA interference reduced neuroblastoma cell proliferation, indicating that HMGA1 is a novel MYCN target gene relevant for neuroblastoma tumorigenesis.
Collapse
Affiliation(s)
- Giuseppe Giannini
- Department of Experimental Medicine and Pathology, University La Sapienza, Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Treff NR, Pouchnik D, Dement GA, Britt RL, Reeves R. High-mobility group A1a protein regulates Ras/ERK signaling in MCF-7 human breast cancer cells. Oncogene 2004; 23:777-85. [PMID: 14737112 DOI: 10.1038/sj.onc.1207167] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
High-mobility group (HMG) A1 proteins are gene regulatory factors whose overexpression is frequently observed in naturally occurring human cancers. The overexpression of transgenic HMGA1 proteins in cells results in neoplastic transformation and promotes progression to malignant cellular phenotypes. To understand the underlying molecular and biological events involved in these phenomena, we used oligonucleotide microarray analyses to generate an HMGA1a-induced expression profile for approximately 22,000 genes. This gene expression profile was generated using a well-characterized transgenic human MCF-7 mammary adenocarcinoma cell line in which overexpression of transgenic HMGA1 promotes a transition to a more malignant and metastatic phenotype. Microarray expression analyses, together with independent quantitative real-time reverse transcriptase polymerase chain reaction results, indicate that HMGA1a regulates genes involved in the Ras-extracellular signal-related kinase (Ras/ERK) mitogenic signaling pathway, including KIT ligand and caveolins 1 and 2. We also found that many cholesterol biosynthesis genes were decreased in cells overexpressing HMGA1a. Cholesterol depletion, decreased caveolin, and increased KIT ligand expression, are all independently associated with the activation of Ras/ERK signaling. Upon further analysis, we found that sensitivity to epidermal growth factor activation of ERK phosphorylation was significantly higher, and that cholesterol was significantly depleted, in cells overexpressing HMGA1a. The cumulative evidence indicates that one likely mechanism by which the HMGA1a protein promotes malignant changes in cells is through increased sensitivity to the activation of the Ras/ERK signaling pathway.
Collapse
Affiliation(s)
- Nathan R Treff
- School of Molecular Biosciences, Washington State University, PO Box 644660, Pullman, WA 99164-4660, USA
| | | | | | | | | |
Collapse
|
32
|
Evans A, Lennard TWJ, Davies BR. High-mobility group protein 1(Y): Metastasis-associated or metastasis-inducing? J Surg Oncol 2004; 88:86-99. [PMID: 15499602 DOI: 10.1002/jso.20136] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Metastasis is the major cause of mortality and morbidity for patients with cancer. The high-mobility group protein 1(Y) [HMG-1(Y)] has a role in the transcription of many genes involved at different steps in the metastatic cascade and has been linked with cancer in human and animal models. This may represent a potential therapeutic target for patients. The following review summarizes and critically appraises the evidence for the role of HMG-1(Y) in metastasis.
Collapse
Affiliation(s)
- Alice Evans
- School of Surgical and Reproductive Sciences and Northern Institute for Cancer Research, Faculty of Medical Sciences, University of Newcastle-upon-Tyne, United Kingdom
| | | | | |
Collapse
|
33
|
Yuza Y, Agawa M, Matsuzaki M, Yamada H, Urashima M. Gene and protein expression profiling during differentiation of neuroblastoma cells triggered by 13-cis retinoic acid. J Pediatr Hematol Oncol 2003; 25:715-20. [PMID: 12972807 DOI: 10.1097/00043426-200309000-00008] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE The precise changes in RNA and protein expression that accompany neuroblastoma differentiation remain unknown. The authors used microarray technologies to screen molecules associated with the differentiation of neuroblastoma (NB) cells induced by 13-cis retinoic acid. METHODS The authors quantified the expression of 2,061 RNA transcripts related to oncogenesis and of 380 proteins expressed in SK-N-SH and CHP-134 NB cell lines in the presence or absence of 13-cis retinoic acid. RESULTS Hierarchical clustering captured gene expression altered during neuroblastoma differentiation induced by 13-cis retinoic acid. Several genes were further abstracted based on P values below 0.0017 or protein chips observed in both NB cell lines. The altered expressions of gene products revealed by both DNA and protein chips were in agreement. The expressions of N-myc, cyclin D3, and Wnt10B were downregulated, whereas those of retinoblastoma (RB) and related genes (p107, RB2/p130, p300/CBP, E2F-1, DP-1) as well as others were upregulated. CONCLUSIONS These results suggest that microarray technology can screen for genes that are important in neuroblastoma differentiation.
Collapse
Affiliation(s)
- Yuki Yuza
- Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan
| | | | | | | | | |
Collapse
|
34
|
Reeves R, Beckerbauer L. HMGI/Y proteins: flexible regulators of transcription and chromatin structure. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1519:13-29. [PMID: 11406267 DOI: 10.1016/s0167-4781(01)00215-9] [Citation(s) in RCA: 285] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The mammalian HMGI/Y (HMGA) non-histone proteins participate in a wide variety of cellular processes including regulation of inducible gene transcription, integration of retroviruses into chromosomes and the induction of neoplastic transformation and promotion of metastatic progression of cancer cells. Recent advances have contributed greatly to our understanding of how the HMGI/Y proteins participate in the molecular mechanisms underlying these biological events. All members of the HMGI/Y family of 'high mobility group' proteins are characterized by the presence of multiple copies of a conserved DNA-binding peptide motif called the 'AT hook' that preferentially binds to the narrow minor groove of stretches of AT-rich sequence. The mammalian HMGI/Y proteins have little, if any, secondary structure in solution but assume distinct conformations when bound to substrates such as DNA or other proteins. Their intrinsic flexibility allows the HMGI/Y proteins to participate in specific protein-DNA and protein-protein interactions that induce both structural changes in chromatin substrates and the formation of stereospecific complexes called 'enhanceosomes' on the promoter/enhancer regions of genes whose transcription they regulate. The formation of such regulatory complexes is characterized by reciprocal inductions of conformational changes in both the HMGI/Y proteins themselves and in their interacting substrates. It may well be that the inherent flexibility of the HMGI/Y proteins, combined with their ability to undergo reversible disordered-to-ordered structural transitions, has been a significant factor in the evolutionary selection of these proteins for their functional role(s) in cells.
Collapse
Affiliation(s)
- R Reeves
- Department of Biochemistry/Biophysics, School of Molecular Biosciences, Washington State University, Pullman, WA 99164-4660, USA.
| | | |
Collapse
|