1
|
Tekin L, Edgünlü T, Genç D. Immunohistochemical and molecular evaluation of TUSC2 expression in breast cancer. Mol Biol Rep 2024; 51:394. [PMID: 38446366 DOI: 10.1007/s11033-024-09320-z] [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: 11/27/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVE Tumor suppressor candidate 2 has shown to be deleted in lung, colon, and bladder cancer types. In the present study, we aimed to investigate the expression of TUSC2 in breast cancer. MATERIALS AND METHODS A total of thirty patients with breast cancer were included in the study. Normal and tumor tissue samples from fresh mastectomy materials were stored at -80 C until the number of cases was completed for gene expression analysis. Histopathological examination was carried out with routine hematoxylin & eosin method. TUSC2 staining was performed for immunohistochemical analysis. RESULTS The tumors of thirteen patients were Luminal A, fourteen patients were Luminal B, one patient was cerbB2(+), and tumors of two patients were triple-negative. Ki67 proliferation index was less than 14% in fifteen cases and tumor size was less than 2 cm in seven cases. Lymphovascular invasion and lymph node metastasis were present in thirteen cases. Statistically, TUSC2 expression significantly decreased or was lost in breast tumor tissues compared to normal tissues (p < 0.0001). TUSC2 expression decreased as the Ki67 proliferation index increased (p = 0.0003), and TUSC2 expression decreased as tumor size increased (p = 0.0483). The loss or decrease in the TUSC2 expression was significant as the tumor grade increased (p = 0.3740). Gene expression analysis correlated with immunohistochemistry results. CONCLUSION The results of the present study demonstrated a decrease or loss of TUSC2 expression in breast cancer tissue compared to normal tissue. A correlation was found between TUSC2 expression and Ki67 proliferation index and tumor size.
Collapse
Affiliation(s)
- Leyla Tekin
- Faculty of Medicine, Pathology Department, Muğla Sıtkı Koçman University, Muğla, Turkey.
| | - Tuba Edgünlü
- Faculty of Medicine, Genetics Department, Muğla Sıtkı Koçman University, Muğla, Turkey
| | - Deniz Genç
- Faculty of Health Sciences, Muğla Sıtkı Koçman University, Muğla, Turkey
| |
Collapse
|
2
|
Arrigo A, Regua AT, Najjar MK, Lo HW. Tumor Suppressor Candidate 2 (TUSC2): Discovery, Functions, and Cancer Therapy. Cancers (Basel) 2023; 15:2455. [PMID: 37173921 PMCID: PMC10177220 DOI: 10.3390/cancers15092455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
Tumor Suppressor Candidate 2 (TUSC2) was first discovered as a potential tumor suppressor gene residing in the frequently deleted 3p21.3 chromosomal region. Since its discovery, TUSC2 has been found to play vital roles in normal immune function, and TUSC2 loss is associated with the development of autoimmune diseases as well as impaired responses within the innate immune system. TUSC2 also plays a vital role in regulating normal cellular mitochondrial calcium movement and homeostasis. Moreover, TUSC2 serves as an important factor in premature aging. In addition to TUSC2's normal cellular functions, TUSC2 has been studied as a tumor suppressor gene that is frequently deleted or lost in a multitude of cancers, including glioma, sarcoma, and cancers of the lung, breast, ovaries, and thyroid. TUSC2 is frequently lost in cancer due to somatic deletion within the 3p21.3 region, transcriptional inactivation via TUSC2 promoter methylation, post-transcriptional regulation via microRNAs, and post-translational regulation via polyubiquitination and proteasomal degradation. Additionally, restoration of TUSC2 expression promotes tumor suppression, eventuating in decreased cell proliferation, stemness, and tumor growth, as well as increased apoptosis. Consequently, TUSC2 gene therapy has been tested in patients with non-small cell lung cancer. This review will focus on the current understanding of TUSC2 functions in both normal and cancerous tissues, mechanisms of TUSC2 loss, TUSC2 cancer therapeutics, open questions, and future directions.
Collapse
Affiliation(s)
- Austin Arrigo
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA (A.T.R.); (M.K.N.)
- Graduate School of Arts and Sciences, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Angelina T. Regua
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA (A.T.R.); (M.K.N.)
| | - Mariana K. Najjar
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA (A.T.R.); (M.K.N.)
- Graduate School of Arts and Sciences, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Hui-Wen Lo
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA (A.T.R.); (M.K.N.)
| |
Collapse
|
3
|
Uzhachenko R, Shimamoto A, Chirwa SS, Ivanov SV, Ivanova AV, Shanker A. Mitochondrial Fus1/Tusc2 and cellular Ca2 + homeostasis: tumor suppressor, anti-inflammatory and anti-aging implications. Cancer Gene Ther 2022; 29:1307-1320. [PMID: 35181743 PMCID: PMC9576590 DOI: 10.1038/s41417-022-00434-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/22/2021] [Accepted: 01/28/2022] [Indexed: 12/02/2022]
Abstract
FUS1/TUSC2 (FUSion1/TUmor Suppressor Candidate 2) is a tumor suppressor gene (TSG) originally described as a member of the TSG cluster from human 3p21.3 chromosomal region frequently deleted in lung cancer. Its role as a TSG in lung, breast, bone, and other cancers was demonstrated by several groups, but molecular mechanisms of its activities are starting to unveil lately. They suggest that Fus1-dependent mechanisms are relevant in etiologies of diseases beyond cancer, such as chronic inflammation, bacterial and viral infections, premature aging, and geriatric diseases. Here, we revisit the discovery of FUS1 gene in the context of tumor initiation and progression, and review 20 years of research into FUS1 functions and its molecular, structural, and biological aspects that have led to its use in clinical trials and gene therapy. We present a data-driven view on how interactions of Fus1 with the mitochondrial Ca2+ (mitoCa2+) transport machinery maintain cellular Ca2+ homeostasis and control cell apoptosis and senescence. This Fus1-mediated cellular homeostasis is at the crux of tumor suppressor, anti-inflammatory and anti-aging activities.
Collapse
Affiliation(s)
- Roman Uzhachenko
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Akiko Shimamoto
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, TN, USA
| | - Sanika S Chirwa
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Sergey V Ivanov
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Alla V Ivanova
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA.
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA.
- Host-Tumor Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
4
|
Rimkus TK, Arrigo AB, Zhu D, Carpenter RL, Sirkisoon S, Doheny D, Regua AT, Wong GL, Manore S, Wagner C, Lin HK, Jin G, Ruiz J, Chan M, Debinski W, Lo HW. NEDD4 degrades TUSC2 to promote glioblastoma progression. Cancer Lett 2022; 531:124-135. [PMID: 35167936 PMCID: PMC8920049 DOI: 10.1016/j.canlet.2022.01.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 12/15/2022]
Abstract
Whether tumor suppressor candidate 2 (TUSC2) plays an important role in glioblastoma (GBM) progression is largely unknown. Whether TUSC2 undergoes polyubiquitination is unknown. Herein, we report that TUSC2 protein expression is reduced/lost in GBM compared to normal brain due to protein destabilization; TUSC2 mRNA is equally expressed in both tissues. NEDD4 E3 ubiquitin ligase polyubiquitinates TUSC2 at residue K71, and the TUSC2-K71R mutant is resistant to NEDD4-mediated proteasomal degradation. Analysis of GBM specimens showed NEDD4 protein is highly expressed in GBM and the level is inversely correlated with TUSC2 protein levels. Furthermore, TUSC2 restoration induces apoptosis and inhibits patient-derived glioma stem cells (PD-GSCs) in vitro and in vivo. Conversely, TUSC2-knockout promotes PD-GSCs in vitro and in vivo. RNA-Seq analysis and subsequent validations showed GBM cells with TUSC2-knockout expressed increased Bcl-xL and were more resistant to apoptosis induced by a Bcl-xL-specific BH3 mimetic. A TUSC2-knockout gene signature created from the RNA-seq data predicts poor patient survival. Together, these findings establish that NEDD4-mediated polyubiquitination is a novel mechanism for TUSC2 degradation in GBM and that TUSC2 loss promotes GBM progression in part through Bcl-xL upregulation.
Collapse
|
5
|
Yang YW, Marrufo A, Chase J, Woodard GA, Jablons DM, Lemjabbar-Alaoui H. Ponatinib is a potential therapeutic approach for malignant pleural mesothelioma. Exp Lung Res 2020; 47:9-25. [PMID: 33107354 DOI: 10.1080/01902148.2020.1836691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Malignant pleural mesothelioma (MPM) is a rare and deadly malignancy. Current MPM therapies remain inadequate, and outcomes are often disappointing. New meaningful therapeutic approaches are urgently needed. Accumulating evidence indicates that the cAbl pathway promotes various tumor-stimulating processes in MPM. In this study, we sought to determine ponatinib's potential utility, a clinically approved and potent cAbl inhibitor, in MPM treatment. MATERIAL AND METHODS Four MPM lines (MSTO211H, H28, H2452, H2052) were treated with ponatinib in vitro, and their growth was assessed. Scratch wound assay was used to investigate the ponatinib effect on cell migration. The expression levels of pAbl and its downstream effectors pCrkL, pAKT, and pSTAT5 were characterized. The in vivo ponatinib effect was evaluated in human MPM cells derived tumor model. RESULTS In all four MPM lines, significant expression levels of phosphorylated cAbl/Arg and pCrkl were observed. Differentially but strongly, ponatinib inhibited the in vitro cell growth and migration of all four MPM line. Western blot analysis showed that the activation of Abl signaling was blocked in the ponatinib-treated MMP lines. In keeping, the cellular levels of pAbl and its downstream effector pCrkL, pAKT, and pSTAT5 were markedly decrease following ponatinib treatment. Moreover, ponatinib treatment amplified the levels of γH2AX in cells denoting increased double-strand DNA breaks levels. Notably, ponatinib treatment reduced in vivo tumor growth and reduced pCrkl and pSTAT5 levels in tumor samples. CONCLUSION Ponatinib may offer a new therapeutic strategy for MPM patients based on cAbl signaling pathway inhibition.
Collapse
Affiliation(s)
- Yi-Wei Yang
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| | - Angelica Marrufo
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| | - Jillian Chase
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| | - Gavitt A Woodard
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| | - David M Jablons
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| | - Hassan Lemjabbar-Alaoui
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
6
|
Mariniello RM, Maria Orlandella F, De Stefano AE, Iervolino PLC, Smaldone G, Luciano N, Cervone N, Munciguerra F, Esposito S, Mirabelli P, Salvatore G. The TUSC2 Tumour Suppressor Inhibits the Malignant Phenotype of Human Thyroid Cancer Cells via SMAC/DIABLO Protein. Int J Mol Sci 2020; 21:ijms21030702. [PMID: 31973107 PMCID: PMC7037188 DOI: 10.3390/ijms21030702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/15/2020] [Indexed: 12/11/2022] Open
Abstract
Thyroid carcinoma is the most common endocrine cancer and includes different forms. Among these, anaplastic thyroid carcinoma (ATC) is the rarest but the most lethal subtype, compared to papillary thyroid carcinoma (PTC) which shows an overall good prognosis. We have previously showed that Tumor Suppressor Candidate 2 (TUSC2), a known tumour suppressor gene, is downregulated in human PTC and ATC compared to normal thyroid samples. The aim of this study was to gain insight into the molecular mechanisms induced by TUSC2 in thyroid cancer cells. Here, we stably transfected TUSC2 in papillary (TPC-1) and in anaplastic (8505C) thyroid cancer cell lines and studied its effects on several biological processes, demonstrating that TUSC2 overexpression decreased thyroid cancer cell proliferation, migration and invasion. Through the proteome profiler apoptosis array, we observed that TUSC2 increased sensitivity to apoptosis by increasing the SMAC/DIABLO and CYTOCHROME C proteins. On the other hand, transient silencing of TUSC2, by siRNA, in an immortalized thyroid follicular epithelial cell line (Nthy-ori 3-1) showed the opposite effect. Finally modulation of SMAC/DIABLO partially rescued the biological effects of TUSC2. Thus, our data highlight a tumour suppressor role of TUSC2 in thyroid carcinogenesis, suggesting that it could be a promising target and biomarker for thyroid carcinoma.
Collapse
Affiliation(s)
- Raffaela Mariarosaria Mariniello
- Dipartimento di Scienze Motorie e del Benessere, Universita’ “Parthenope”, Via Medina 40, 80133 Napoli, Italy
- CEINGE—Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Napoli, Italy
| | | | - Anna Elisa De Stefano
- Dipartimento di Scienze Motorie e del Benessere, Universita’ “Parthenope”, Via Medina 40, 80133 Napoli, Italy
- CEINGE—Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Napoli, Italy
| | - Paola Lucia Chiara Iervolino
- CEINGE—Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Napoli, Italy
- Dipartimento di Scienze Biomediche Avanzate, Universita’ “Federico II”, Via Pansini 5, 80131 Napoli, Italy
| | | | - Neila Luciano
- CEINGE—Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Napoli, Italy
| | - Nara Cervone
- CEINGE—Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Napoli, Italy
| | - Francesco Munciguerra
- CEINGE—Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Napoli, Italy
| | - Silvia Esposito
- CEINGE—Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Napoli, Italy
| | | | - Giuliana Salvatore
- Dipartimento di Scienze Motorie e del Benessere, Universita’ “Parthenope”, Via Medina 40, 80133 Napoli, Italy
- CEINGE—Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Napoli, Italy
- IRCCS SDN, Via Emanuele Gianturco 113, 80143 Napoli, Italy
- Correspondence:
| |
Collapse
|
7
|
Shen X, Zhang L, Li J, Li Y, Wang Y, Xu ZX. Recent Findings in the Regulation of Programmed Death Ligand 1 Expression. Front Immunol 2019; 10:1337. [PMID: 31258527 PMCID: PMC6587331 DOI: 10.3389/fimmu.2019.01337] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022] Open
Abstract
With the recent approvals for the application of monoclonal antibodies that target the well-characterized immune checkpoints, immune therapy shows great potential against both solid and hematologic tumors. The use of these therapeutic monoclonal antibodies elicits inspiring clinical results with durable objective responses and improvements in overall survival. Agents targeting programmed cell death protein 1 (PD-1; also known as PDCD1) and its ligand (PD-L1) achieve a great success in immune checkpoints therapy. However, the majority of patients fail to respond to PD-1/PD-L1 axis inhibitors. Expression of PD-L1 on the membrane of tumor and immune cells has been shown to be associated with enhanced objective response rates to PD-1/PD-L1 inhibition. Thus, an improved understanding of how PD-L1 expression is regulated will enable us to better define its role as a predictive marker. In this review, we summarize recent findings in the regulation of PD-L1 expression.
Collapse
Affiliation(s)
- Xiangfeng Shen
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Lihong Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Jicheng Li
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yulin Li
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China
| |
Collapse
|
8
|
Chen J, Song B, Kong G. MicroRNA‑663b downregulation inhibits proliferation and induces apoptosis in bladder cancer cells by targeting TUSC2. Mol Med Rep 2019; 19:3896-3902. [PMID: 30896880 DOI: 10.3892/mmr.2019.10023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 01/25/2019] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to explore the role and underlying molecular mechanism of microRNA‑663b (miR‑663b) in the tumorigenesis of bladder cancer. The miR‑663b expression in human bladder cancer tissues and cell lines was measured determined reverse transcription‑quantitative polymerase chain reaction. TargetScan was used to predict the potential targets of miR‑663b, and a dual‑luciferase reporter assay was performed to validate tumor suppressor candidate 2 (TUSC2) as a target of miR‑663b. Cell Counting Kit‑8 was used for cell viability analysis, and cell apoptosis was evaluated by flow cytometry. In addition, western blot analysis was performed to detect protein expression in current study. The findings suggested that miR‑663b was upregulated in bladder cancer tissues and cell lines compared with normal tissue and cells. TUSC2 was validated as a direct target of miR‑663b and was negatively regulated by miR‑663b. miR‑663b inhibition significantly reduced the viability of T24 cells, and T24 cell apoptosis was markedly induced. In addition, miR‑663b inhibition enhanced the expression levels of p53 and p21 in T24 cells. Furthermore, the changes caused by miR‑663b inhibitor in T24 cells were eliminated by TUSC2 gene silencing. In conclusion, inhibition of miR‑663b reduced viability and induced apoptosis in bladder cancer cells by targeting TUSC2. These findings provide a promising novel therapeutic target for bladder cancer treatment.
Collapse
Affiliation(s)
- Jianying Chen
- Urology Department, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Bo Song
- Urology Department, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Guangqi Kong
- Urology Department, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, P.R. China
| |
Collapse
|
9
|
Kim I, Kim JH, Kim K, Seong S, Kim N. Tusc2/Fus1 regulates osteoclast differentiation through NF-κB and NFATc1. BMB Rep 2018; 50:454-459. [PMID: 28391779 PMCID: PMC5625692 DOI: 10.5483/bmbrep.2017.50.9.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Indexed: 01/20/2023] Open
Abstract
Tumor suppressor candidate 2 (Tusc2, also known as Fus1) regulates calcium signaling, and Ca2+-dependent nuclear factor of activated T-cells (NFAT) and nuclear factor kappa B (NF-κB) pathways, which play roles in osteoclast differentiation. However, the role of Tusc2 in osteoclasts remains unknown. Here, we report that Tusc2 positively regulates the differentiation of osteoclasts. Overexpression of Tusc2 in osteoclast precursor cells enhanced receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclast differentiation. In contrast, small interfering RNA-mediated knockdown of Tusc2 strongly inhibited osteoclast differentiation. In addition, Tusc2 induced the activation of RANKL-mediated NF-κB and calcium/calmodulin-dependent kinase IV (CaMKIV)/cAMP-response element (CRE)-binding protein CREB signaling cascades. Taken together, these results suggest that Tusc2 acts as a positive regulator of RANKL-mediated osteoclast differentiation.
Collapse
Affiliation(s)
- Inyoung Kim
- Departments of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Jung Ha Kim
- Departments of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Kabsun Kim
- Departments of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Semun Seong
- Departments of Pharmacology and Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Nacksung Kim
- Departments of Pharmacology and Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea
| |
Collapse
|
10
|
Tang X, Zhou J, Zhang J, Zhou LY, Zhai LL, Vanessa MED, Yi J, Yi YY, Lin J, Deng ZQ. Low Expression of FUS1 Is Negatively Correlated with miR-378 and May Predict Adverse Prognoses in Acute Myeloid Leukemia. Acta Haematol 2018; 139:89-95. [PMID: 29393096 DOI: 10.1159/000486663] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/23/2017] [Indexed: 12/28/2022]
Abstract
FUS1 is a tumor suppressor gene that has been found to be frequently lost in a variety of solid tumors. In this study, we aimed to investigate the expression status of the FUS1 gene in acute myeloid leukemia (AML), as well as its clinical significance. We further explored the correlation between the expression of FUS1 and miR-378 in AML. We detected expression of the FUS1 transcript in bone marrow mononuclear cells from 23 controls and 158 newly diagnosed AML patients by real-time quantitative polymerase chain reaction. Downregulated FUS1 expression was found in 139 out of 158 (87.97%) AML cases; this rate was significantly lower than that in all 23 controls (p = 0.012). Receiver operating characteristic curve analysis revealed that the FUS1 transcript level could discriminate AML patients from controls effectively (area under the ROC curve = 0.663). Kaplan-Meier analysis demonstrated that non-M3-AML patients with a low FUS1 expression had a shorter overall survival (p = 0.049) and leukemia-free survival (p = 0.051) than those with a high FUS1 expression. Furthermore, we studied the correlation between the expression of FUS1 and miR-378 in 53 newly diagnosed AML patients. We found that the correlation coefficient was -0.346, which showed that FUS1 and miR-378 were negatively correlated in AML patients (p = 0.011). These results indicate that the low expression of FUS1 is a common molecular event in AML.
Collapse
Affiliation(s)
- Xi Tang
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jiao Zhou
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jing Zhang
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Ling-Yu Zhou
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Ling-Ling Zhai
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Minse Evola-Deniz Vanessa
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jing Yi
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Yun-Yun Yi
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jiang Lin
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Zhao-Qun Deng
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
11
|
Cao X, Zhao Y, Wang J, Dai B, Gentile E, Lin J, Pu X, Ji L, Wu S, Meraz I, Majidi M, Roth JA. TUSC2 downregulates PD-L1 expression in non-small cell lung cancer (NSCLC). Oncotarget 2017; 8:107621-107629. [PMID: 29296193 PMCID: PMC5746095 DOI: 10.18632/oncotarget.22581] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/01/2017] [Indexed: 12/21/2022] Open
Abstract
Expression of the TUSC2 tumor-suppressor gene in TUSC2-deficient NSCLC cells decreased PD-L1 expression and inhibited mTOR activity. Overexpressing TUSC2 or treatment with rapamycin resulted in similar inhibition of PD-L1 expression. Both TUSC2 and rapamycin decreased p70 and SK6 phosphorylation, suggesting that TUSC2 and rapamycin share the same mTOR target. Microarray mRNA expression analysis using TUSC2-inducible H1299 showed that genes that negatively regulate the mTOR pathway were significantly upregulated by TUSC2 compared with control. The presence of IFN-γ significantly increased PD-L1 expression in lung cancer cell lines, but overexpressing TUSC2 in these cell lines prevented PD-L1 from increasing in the presence of IFN-γ. Taken together, these findings show that TUSC2 can decrease PD-L1 expression in lung cancer cells. This ability to modify the tumor microenvironment suggests that TUSC2 could be added to checkpoint inhibitors to improve the treatment of lung cancer.
Collapse
Affiliation(s)
- Xiaobo Cao
- Department of Thoracic and Cardiovascular Surgery, Section of Thoracic Molecular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yang Zhao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bingbing Dai
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Emanuela Gentile
- Department of Thoracic and Cardiovascular Surgery, Section of Thoracic Molecular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jing Lin
- Department of Thoracic and Cardiovascular Surgery, Section of Thoracic Molecular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xingxiang Pu
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital, Changsha, China
| | - Lin Ji
- Department of Thoracic and Cardiovascular Surgery, Section of Thoracic Molecular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shuhong Wu
- Department of Thoracic and Cardiovascular Surgery, Section of Thoracic Molecular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ismail Meraz
- Department of Thoracic and Cardiovascular Surgery, Section of Thoracic Molecular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mourad Majidi
- Department of Thoracic and Cardiovascular Surgery, Section of Thoracic Molecular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, Section of Thoracic Molecular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
12
|
Uzhachenko R, Boyd K, Olivares-Villagomez D, Zhu Y, Goodwin JS, Rana T, Shanker A, Tan WJT, Bondar T, Medzhitov R, Ivanova AV. Mitochondrial protein Fus1/Tusc2 in premature aging and age-related pathologies: critical roles of calcium and energy homeostasis. Aging (Albany NY) 2017; 9:627-649. [PMID: 28351997 PMCID: PMC5391223 DOI: 10.18632/aging.101213] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/18/2017] [Indexed: 12/20/2022]
Abstract
Decreased energy production and increased oxidative stress are considered to be major contributors to aging and aging-associated pathologies. The role of mitochondrial calcium homeostasis has also been highlighted as an important factor affecting different pathological conditions. Here, we present evidence that loss of a small mitochondrial protein Fus1 that maintains mitochondrial homeostasis results in premature aging, aging-associated pathologies, and decreased survival. We showed that Fus1KO mice develop multiple early aging signs including lordokyphosis, lack of vigor, inability to accumulate fat, reduced ability to tolerate stress, and premature death. Other prominent pathological changes included low sperm counts, compromised ability of adult stem cells to repopulate tissues, and chronic inflammation. At the molecular level, we demonstrated that mitochondria of Fus1 KO cells have low reserve respiratory capacity (the ability to produce extra energy during sudden energy demanding situations), and show significantly altered dynamics of cellular calcium response. Our recent studies on early hearing and memory loss in Fus1 KO mice combined with the new data presented here suggest that calcium and energy homeostasis controlled by Fus1 may be at the core of its aging-regulating activities. Thus, Fus1 protein and Fus1-dependent pathways and processes may represent new tools and targets for anti-aging strategies.
Collapse
Affiliation(s)
- Roman Uzhachenko
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Kelli Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Danyvid Olivares-Villagomez
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Yueming Zhu
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - J Shawn Goodwin
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Tanu Rana
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA.,Present address: Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37235, USA
| | - Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA.,Department of Surgery, Section of Otolaryngology, Yale University School of Medicine, New Haven, CT 0651, USA
| | - Winston J T Tan
- Department of Surgery, Section of Otolaryngology, Yale University School of Medicine, New Haven, CT 0651, USA
| | - Tanya Bondar
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 0651, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 0651, USA
| | - Alla V Ivanova
- Department of Surgery, Section of Otolaryngology, Yale University School of Medicine, New Haven, CT 0651, USA
| |
Collapse
|
13
|
Tumor Suppressor microRNAs Contribute to the Regulation of PD-L1 Expression in Malignant Pleural Mesothelioma. J Thorac Oncol 2017. [DOI: 10.1016/j.jtho.2017.05.024] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
14
|
Rimkus T, Sirkisoon S, Harrison A, Lo HW. Tumor suppressor candidate 2 (TUSC2, FUS-1) and human cancers. DISCOVERY MEDICINE 2017; 23:325-330. [PMID: 28715648 PMCID: PMC5808457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Tumor suppressor candidate 2 (TUSC2, also known as FUS1) was identified in 2000 as a candidate tumor suppressor gene located in a region on chromosome 3p21.3 that is homozygously deleted in some lung and breast cancers. The deletion is rare in lung and breast cancers, but is frequent in malignant pleural mesothelioma. Evidence to date indicates that TUSC2 behaves as a tumor suppressor in lung cancer; however, its role as a tumor suppressor for other tumor types has not been fully established. Loss of TUSC2 expression at the mRNA and protein levels has been reported in various cancers. While the mechanisms underlying the loss are still not well understood, several microRNAs have been reported to downregulate TUSC2 expression. TUSC2 elicits its anti-tumor effects through regulating G1 cell cycle progression, apoptosis, calcium homeostasis, gene expression, and the activity of various protein tyrosine kinases and Ser/Thr kinases, albeit the precise mechanisms that TUSC2 utilizes to regulate these cellular processes and signaling molecules are still elusive. TUSC2 restoration has been exploited as an anti-cancer therapy in various cancers in preclinical models, and clinically in patients with lung cancer. The first-in-human phase I trial demonstrated desirable safety outcomes. Phase I/II trials are being conducted to evaluate the efficacy of combining TUSC2-nanoparticles with erlotinib, an FDA-approved EGFR inhibitor. This review summarizes recent findings that advanced our understanding of TUSC2 as a novel tumor suppressor and a therapeutic opportunity for treating TUSC2-deficient cancers.
Collapse
Affiliation(s)
- Tadas Rimkus
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Sherona Sirkisoon
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Alexandra Harrison
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Hui-Wen Lo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
- Wake Forest Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
15
|
Coronas-Samano G, Baker KL, Tan WJT, Ivanova AV, Verhagen JV. Fus1 KO Mouse As a Model of Oxidative Stress-Mediated Sporadic Alzheimer's Disease: Circadian Disruption and Long-Term Spatial and Olfactory Memory Impairments. Front Aging Neurosci 2016; 8:268. [PMID: 27895577 PMCID: PMC5108791 DOI: 10.3389/fnagi.2016.00268] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 10/24/2016] [Indexed: 12/13/2022] Open
Abstract
Insufficient advances in the development of effective therapeutic treatments of sporadic Alzheimer's Disease (sAD) to date are largely due to the lack of sAD-relevant animal models. While the vast majority of models do recapitulate AD's hallmarks of plaques and tangles by virtue of tau and/or beta amyloid overexpression, these models do not reflect the fact that in sAD (unlike familial AD) these genes are not risk factors per se and that other mechanisms like oxidative stress, metabolic dysregulation and inflammation play key roles in AD etiology. Here we characterize and propose the Fus1 KO mice that lack a mitochondrial protein Fus1/Tusc2 as a new sAD model. To establish sAD relevance, we assessed sAD related deficits in Fus1 KO and WT adult mice of 4-5 months old, the equivalent human age when the earliest cognitive and olfactory sAD symptoms arise. Fus1 KO mice showed oxidative stress (increased levels of ROS, decreased levels of PRDX1), disruption of metabolic homeostasis (decreased levels of ACC2, increased phosphorylation of AMPK), autophagy (decreased levels of LC3-II), PKC (decreased levels of RACK1) and calcium signaling (decreased levels of Calb2) in the olfactory bulb and/or hippocampus. Mice were behaviorally tested using objective and accurate video tracking (Noldus), in which Fus1 KO mice showed clear deficits in olfactory memory (decreased habituation/cross-habituation in the short and long term), olfactory guided navigation memory (inability to reduce their latency to find the hidden cookie), spatial memory (learning impairments on finding the platform in the Morris water maze) and showed more sleep time during the diurnal cycle. Fus1 KO mice did not show clear deficits in olfactory perception (cross-habituation), association memory (passive avoidance) or in species-typical behavior (nest building) and no increased anxiety (open field, light-dark box) or depression/anhedonia (sucrose preference) at this relatively young age. These neurobehavioral deficits of the Fus1 KO mice at this relatively young age are highly relevant to sAD, making them suitable for effective research on pharmacological targets in the context of early intervention of sAD.
Collapse
Affiliation(s)
| | - Keeley L Baker
- The John B. Pierce LaboratoryNew Haven, CT, USA; Department of Neuroscience, Yale University School of MedicineNew Haven, CT, USA
| | - Winston J T Tan
- Department of Surgery, Yale University School of Medicine New Haven, CT, USA
| | - Alla V Ivanova
- Department of Surgery, Yale University School of Medicine New Haven, CT, USA
| | - Justus V Verhagen
- The John B. Pierce LaboratoryNew Haven, CT, USA; Department of Neuroscience, Yale University School of MedicineNew Haven, CT, USA
| |
Collapse
|
16
|
Mitochondria, calcium, and tumor suppressor Fus1: At the crossroad of cancer, inflammation, and autoimmunity. Oncotarget 2016; 6:20754-72. [PMID: 26246474 PMCID: PMC4673227 DOI: 10.18632/oncotarget.4537] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/27/2015] [Indexed: 12/12/2022] Open
Abstract
Mitochondria present a unique set of key intracellular functions such as ATP synthesis, production of reactive oxygen species (ROS) and Ca2+ buffering. Mitochondria both encode and decode Ca2+ signals and these interrelated functions have a direct impact on cell signaling and metabolism. High proliferative potential is a key energy-demanding feature shared by cancer cells and activated T lymphocytes. Switch of a metabolic state mediated by alterations in mitochondrial homeostasis plays a fundamental role in maintenance of the proliferative state. Recent studies show that tumor suppressors have the ability to affect mitochondrial homeostasis controlling both cancer and autoimmunity. Herein, we discuss established and putative mechanisms of calcium–dependent regulation of both T cell and tumor cell activities. We use the mitochondrial protein Fus1 as a case of tumor suppressor that controls immune response and tumor growth via maintenance of mitochondrial homeostasis. We focus on the regulation of mitochondrial Ca2+ handling as a key function of Fus1 and highlight the mechanisms of a crosstalk between Ca2+ accumulation and mitochondrial homeostasis. Given the important role of Ca2+ signaling, mitochondrial Ca2+ transport and ROS production in the activation of NFAT and NF-κB transcription factors, we outline the importance of Fus1 activities in this context.
Collapse
|
17
|
Patel SJ, Dao S, Darie CC, Clarkson BD. Defective quorum sensing of acute lymphoblastic leukemic cells: evidence of collective behavior of leukemic populations as semi-autonomous aberrant ecosystems. Am J Cancer Res 2016; 6:1177-230. [PMID: 27429840 PMCID: PMC4937729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/04/2016] [Indexed: 06/06/2023] Open
Abstract
Quorum sensing (QS) is a generic term used to describe cell-cell communication and collective decision making by bacterial and social insects to regulate the expression of specific genes in controlling cell density and other properties of the populations in response to nutrient supply or changes in the environment. QS mechanisms also have a role in higher organisms in maintaining homeostasis, regulation of the immune system and collective behavior of cancer cell populations. In the present study, we used a p190(BCR-ABL) driven pre-B acute lymphoblastic leukemia (ALL3) cell line derived from the pleural fluid of a terminally ill patient with ALL to test the QS hypothesis in leukemia. ALL3 cells don't grow at low density (LD) in liquid media but grow progressively faster at increasingly high cell densities (HD) in contrast to other established leukemic cell lines that grow well at very low starting cell densities. The ALL3 cells at LD are poised to grow but shortly die without additional stimulation. Supernates of ALL3 cells (HDSN) and some other primary cells grown at HD stimulate the growth of the LD ALL3 cells without which they won't survive. To get further insight into the activation processes we performed microarray analysis of the LD ALL3 cells after stimulation with ALL3 HDSN at days 1, 3, and 6. This screen identified several candidate genes, and we linked them to signaling networks and their functions. We observed that genes involved in lipid, cholesterol, fatty acid metabolism, and B cell activation are most up- or down-regulated upon stimulation of the LD ALL3 cells using HDSN. We also discuss other pathways that are differentially expressed upon stimulation of the LD ALL3 cells. Our findings suggest that the Ph+ ALL population achieves dominance by functioning as a collective aberrant ecosystem subject to defective quorum-sensing regulatory mechanisms.
Collapse
Affiliation(s)
- Sapan J Patel
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program1275 York Avenue, Box #96, New York, NY 10065, USA
- Department of Chemistry and Biomolecular Science, Biochemistry and Proteomics Group, Clarkson University8 Clarkson Avenue, Potsdam, NY 13699-5810, USA
| | - Su Dao
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program1275 York Avenue, Box #96, New York, NY 10065, USA
| | - Costel C Darie
- Department of Chemistry and Biomolecular Science, Biochemistry and Proteomics Group, Clarkson University8 Clarkson Avenue, Potsdam, NY 13699-5810, USA
| | - Bayard D Clarkson
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program1275 York Avenue, Box #96, New York, NY 10065, USA
| |
Collapse
|
18
|
Cellular oxidative stress response mediates radiosensitivity in Fus1-deficient mice. Cell Death Dis 2015; 6:e1652. [PMID: 25695605 PMCID: PMC4669799 DOI: 10.1038/cddis.2014.593] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 12/08/2014] [Accepted: 12/19/2014] [Indexed: 01/26/2023]
Abstract
Mechanism of radiosensitivity of normal tissues, a key factor in determining the toxic side effects of cancer radiotherapy, is not fully understood. We recently demonstrated that deficiency of mitochondrial tumor suppressor, Fus1, increases radiosensitivity at the organismal, tissue and cellular levels. Since Fus1-deficient mice and cells exhibit high levels of oxidative stress, we hypothesized that dysregulation of cellular antioxidant defenses may contribute to the increased radiosensitivity. To address this potential mechanism, we treated the Fus1 KO mice with an inhibitor of pathogenic oxidative reactions, pyridoxamine (PM). Treatment with PM ameliorated IR-induced damage to GI epithelium of Fus1 KO mice and significantly increased the survival of irradiated mice. In cultured Fus1 KO epithelial cells, IR-induced oxidative stress was enhanced because of inadequate cellular antioxidant defenses, such as low levels and/or activities of cytochrome C, Sod 2 and STAT3. This resulted in dysregulation of IR-induced DNA-damage response and DNA synthesis. Treatment of Fus1 KO cells with PM or Sod 2 mimetic Tempol normalized the oxidative stress response, thus compensating to a significant degree for inadequate antioxidant response. Our findings using Fus1 KO radiosensitive mice suggest that radiosensitivity is mediated via dysregulation of antioxidant response and defective redox homeostasis.
Collapse
|
19
|
Uzhachenko R, Ivanov SV, Yarbrough WG, Shanker A, Medzhitov R, Ivanova AV. Fus1/Tusc2 is a novel regulator of mitochondrial calcium handling, Ca2+-coupled mitochondrial processes, and Ca2+-dependent NFAT and NF-κB pathways in CD4+ T cells. Antioxid Redox Signal 2014; 20:1533-47. [PMID: 24328503 PMCID: PMC3942676 DOI: 10.1089/ars.2013.5437] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS Fus1 has been established as mitochondrial tumor suppressor, immunomodulator, and antioxidant protein, but molecular mechanism of these activities remained to be identified. Based on putative calcium-binding and myristoyl-binding domains that we identified in Fus1, we explored our hypothesis that Fus1 regulates mitochondrial calcium handling and calcium-coupled processes. RESULTS Fus1 loss resulted in reduced rate of mitochondrial calcium uptake in calcium-loaded epithelial cells, splenocytes, and activated CD4(+) T cells. The reduced rate of mitochondrial calcium uptake in Fus1-deficient cells correlated with cytosolic calcium increase and dysregulation of calcium-coupled mitochondrial parameters, such as reactive oxygen species production, ΔμH(+), mitochondrial permeability transition pore opening, and GSH content. Inhibition of calcium efflux via mitochondria, Na(+)/Ca(2+) exchanger significantly improved the mitochondrial calcium uptake in Fus1(-/-) cells. Ex vivo analysis of activated CD4(+) T cells showed Fus1-dependent changes in calcium-regulated processes, such as surface expression of CD4 and PD1/PD-L1, proliferation, and Th polarization. Fus1(-/-) T cells showed increased basal expression of calcium-dependent NF-κB and NFAT targets but were unable to fully activate these pathways after stimulation. INNOVATION Our results establish Fus1 as one of the few identified regulators of mitochondrial calcium handling. Our data support the idea that alterations in mitochondrial calcium dynamics could lead to the disruption of metabolic coupling in mitochondria that, in turn, may result in multiple cellular and systemic abnormalities. CONCLUSION Our findings suggest that Fus1 achieves its protective role in inflammation, autoimmunity, and cancer via the regulation of mitochondrial calcium and calcium-coupled parameters.
Collapse
Affiliation(s)
- Roman Uzhachenko
- 1 Department of Biochemistry and Cancer Biology, VICC, Meharry Medical College , Nashville, Tennessee
| | | | | | | | | | | |
Collapse
|
20
|
Zhang C, Kho YS, Wang Z, Chiang YT, Ng GKH, Shaw PC, Wang Y, Qi RZ. Transmembrane and coiled-coil domain family 1 is a novel protein of the endoplasmic reticulum. PLoS One 2014; 9:e85206. [PMID: 24454821 PMCID: PMC3891740 DOI: 10.1371/journal.pone.0085206] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/23/2013] [Indexed: 01/01/2023] Open
Abstract
The endoplasmic reticulum (ER) is a continuous membrane network in eukaryotic cells comprising the nuclear envelope, the rough ER, and the smooth ER. The ER has multiple critical functions and a characteristic structure. In this study, we identified a new protein of the ER, TMCC1 (transmembrane and coiled-coil domain family 1). The TMCC family consists of at least 3 putative proteins (TMCC1-3) that are conserved from nematode to human. We show that TMCC1 is an ER protein that is expressed in diverse human cell lines. TMCC1 contains 2 adjacent transmembrane domains near the C-terminus, in addition to coiled-coil domains. TMCC1 was targeted to the rough ER through the transmembrane domains, whereas the N-terminal region and C-terminal tail of TMCC1 were found to reside in the cytoplasm. Moreover, the cytosolic region of TMCC1 formed homo- or hetero-dimers or oligomers with other TMCC proteins and interacted with ribosomal proteins. Notably, overexpression of TMCC1 or its transmembrane domains caused defects in ER morphology. Our results suggest roles of TMCC1 in ER organization.
Collapse
Affiliation(s)
- Chao Zhang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yik-Shing Kho
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Zhe Wang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yan Ting Chiang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Department of Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Gary K. H. Ng
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Pang-Chui Shaw
- Biochemistry Programme and Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Robert Z. Qi
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- * E-mail:
| |
Collapse
|
21
|
Meng J, Majidi M, Fang B, Ji L, Bekele BN, Minna JD, Roth JA. The tumor suppressor gene TUSC2 (FUS1) sensitizes NSCLC to the AKT inhibitor MK2206 in LKB1-dependent manner. PLoS One 2013; 8:e77067. [PMID: 24146957 PMCID: PMC3798310 DOI: 10.1371/journal.pone.0077067] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/29/2013] [Indexed: 12/20/2022] Open
Abstract
TUSC2-defective gene expression is detected in the majority of lung cancers and is associated with worse overall survival. We analyzed the effects of TUSC2 re-expression on tumor cell sensitivity to the AKT inhibitor, MK2206, and explored their mutual signaling connections, in vitro and in vivo. TUSC2 transient expression in three LKB1-defective non-small cell lung cancer (NSCLC) cell lines combined with MK2206 treatment resulted in increased repression of cell viability and colony formation, and increased apoptotic activity. In contrast, TUSC2 did not affect the response to MK2206 treatment for two LKB1-wild type NSCLC cell lines. In vivo, TUSC2 systemic delivery, by nanoparticle gene transfer, combined with MK2206 treatment markedly inhibited growth of tumors in a human LKB1-defective H322 lung cancer xenograft mouse model. Biochemical analysis showed that TUSC2 transient expression in LKB1-defective NSCLC cells significantly stimulated AMP-activated protein kinase (AMPK) phosphorylation and enzymatic activity. More importantly, AMPK gene knockdown abrogated TUSC2-MK2206 cooperation, as evidenced by reduced sensitivity to the combined treatment. Together, TUSC2 re-expression and MK2206 treatment was more effective in inhibiting the phosphorylation and kinase activities of AKT and mTOR proteins than either single agent alone. In conclusion, these findings support the hypothesis that TUSC2 expression status is a biological variable that potentiates MK2206 sensitivity in LKB1-defective NSCLC cells, and identifies the AMPK/AKT/mTOR signaling axis as an important regulator of this activity.
Collapse
Affiliation(s)
- Jieru Meng
- Section of Thoracic Molecular Oncology, Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| | - Mourad Majidi
- Section of Thoracic Molecular Oncology, Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Bingliang Fang
- Section of Thoracic Molecular Oncology, Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Lin Ji
- Section of Thoracic Molecular Oncology, Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - B. Nebiyou Bekele
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - John D. Minna
- Hamon Center for Therapeutic Oncology Research and Simmons Cancer Center, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Jack A. Roth
- Section of Thoracic Molecular Oncology, Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
22
|
Loss of mitochondrial protein Fus1 augments host resistance to Acinetobacter baumannii infection. Infect Immun 2013; 81:4461-9. [PMID: 24042119 DOI: 10.1128/iai.00771-13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Fus1 is a tumor suppressor protein with recently described immunoregulatory functions. Although its role in sterile inflammation is being elucidated, its role in regulating immune responses to infectious agents has not been examined. We used here a murine model of Acinetobacter baumannii pneumonia to identify the role of Fus1 in antibacterial host defenses. We found that the loss of Fus1 in mice results in significantly increased resistance to A. baumannii pneumonia. We observed earlier and more robust recruitment of neutrophils and macrophages to the lungs of infected Fus1(-/-) mice, with a concomitant increase in phagocytosis of invading bacteria and more rapid clearance. Such a prompt and enhanced immune response to bacterial infection in Fus1(-/-) mice stems from early activation of proinflammatory pathways (NF-κB and phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin [mTOR]), most likely due to significantly increased mitochondrial membrane potential and mitochondrial reactive oxygen species production. Significant early upregulation of interleukin-17 (IL-17) in Fus1(-/-) immune cells was also observed, together with significant downregulation of IL-10. Depletion of neutrophils eliminates the enhanced antibacterial defenses of the Fus1(-/-) mice, suggesting that ultimately it is the enhanced immune cell recruitment that mediates the increased resistance of Fus1(-/-) mice to A. baumannii pneumonia. Taken together, our data define the novel role for Fus1 in the immune response to A. baumannii pneumonia and highlight new avenues for immune modulating therapeutic targets for this treatment-resistant nosocomial pathogen.
Collapse
|
23
|
Abstract
FUS1/TUSC2 is a mitochondrial tumor suppressor with activity to regulate cellular oxidative stress by maintaining balanced ROS production and mitochondrial homeostasis. Fus1 expression is inhibited by ROS, suggesting that individuals with a high level of ROS may have lower Fus1 in normal tissues and, thus, may be more prone to oxidative stress-induced side effects of cancer treatment, including radiotherapy. As the role of Fus1 in the modulation of cellular radiosensitivity is unknown, we set out to determine molecular mechanisms of Fus1 involvement in the IR response in normal tissues. Mouse whole-body irradiation methodology was employed to determine the role for Fus1 in the radiation response and explore underlying molecular mechanisms. Fus1(-/-) mice were more susceptible to radiation compared with Fus1(+/+) mice, exhibiting increased mortality and accelerated apoptosis of the GI crypt epithelial cells. Following untimely reentrance into the cell cycle, the Fus1(-/-) GI crypt cells died at accelerated rate via mitotic catastrophe that resulted in diminished and/or delayed crypt regeneration after irradiation. At the molecular level, dysregulated dynamics of activation of main IR response proteins (p53, NFκB, and GSK-3β), as well as key signaling pathways involved in oxidative stress response (SOD2, PRDX1, and cytochrome c), apoptosis (BAX and PARP1), cell cycle (Cyclins B1 and D1), and DNA repair (γH2AX) were found in Fus1(-/-) cells after irradiation. Increased radiosensitivity of other tissues, such as immune cells and hair follicles was also detected in Fus1(-/-) mice. Our findings demonstrate a previously unknown radioprotective function of the mitochondrial tumor suppressor Fus1 in normal tissues and suggest new individualized therapeutic approaches based on Fus1 expression.
Collapse
|
24
|
Mossman BT, Shukla A, Heintz NH, Verschraegen CF, Thomas A, Hassan R. New insights into understanding the mechanisms, pathogenesis, and management of malignant mesotheliomas. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1065-77. [PMID: 23395095 DOI: 10.1016/j.ajpath.2012.12.028] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 12/04/2012] [Accepted: 12/24/2012] [Indexed: 12/20/2022]
Abstract
Malignant mesothelioma (MM) is a relatively rare but devastating tumor that is increasing worldwide. Yet, because of difficulties in early diagnosis and resistance to conventional therapies, MM remains a challenge for pathologists and clinicians to treat. In recent years, much has been revealed regarding the mechanisms of interactions of pathogenic fibers with mesothelial cells, crucial signaling pathways, and genetic and epigenetic events that may occur during the pathogenesis of these unusual, pleiomorphic tumors. These observations support a scenario whereby mesothelial cells undergo a series of chronic injury, inflammation, and proliferation in the long latency period of MM development that may be perpetuated by durable fibers, the tumor microenvironment, and inflammatory stimuli. One culprit in sustained inflammation is the activated inflammasome, a component of macrophages or mesothelial cells that leads to production of chemotactic, growth-promoting, and angiogenic cytokines. This information has been vital to designing novel therapeutic approaches for patients with MM that focus on immunotherapy, targeting growth factor receptors and pathways, overcoming resistance to apoptosis, and modifying epigenetic changes.
Collapse
Affiliation(s)
- Brooke T Mossman
- Department of Pathology, University of Vermont College of Medicine, Burlington, Vermont 05405-0068, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Demokan S, Chuang AY, Chang X, Khan T, Smith IM, Pattani KM, Dasgupta S, Begum S, Khan Z, Liegeois NJ, Westra WH, Sidransky D, Koch W, Califano JA. Identification of guanine nucleotide-binding protein γ-7 as an epigenetically silenced gene in head and neck cancer by gene expression profiling. Int J Oncol 2013; 42:1427-36. [PMID: 23403885 PMCID: PMC3981008 DOI: 10.3892/ijo.2013.1808] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 12/10/2012] [Indexed: 01/05/2023] Open
Abstract
Silencing of tumor suppressor genes plays a vital role in head and neck carcinogenesis. Aberrant hypermethylation in the promoter region of some known or putative tumor suppressor genes occurs frequently during the development of various types of cancer including head and neck squamous cell carcinoma (HNSCC). In this study we used an expanded mRNA expression profiling approach followed by microarray expression analysis to identify epigenetically inactivated genes in HNSCC. Two HNSCC cell lines were treated with 5-aza-2'-deoxycytidine followed by microarray analysis to identify epigenetically silenced genes in HNSCC. We found 1,960, 614 and 427 genes were upregulated in the HNSCC cell lines JHU-012, JHU-011 and the combination of both cell lines, respectively. HNSCC tumor and normal mucosal samples were used for gene profiling by a 47K mRNA gene expression array and we found 7,140 genes were downregulated in HNSCC tumors compared to normal mucosa, as determined by microarray analysis, and were integrated with cell line data. Integrative analysis defined 126 candidate genes, of which only seven genes showed differential methylation in tumors and no methylation in normal mucosa after bisulfite sequencing. Following validation by QMSP, one gene, guanine nucleotide-binding protein γ-7 (GNG7), was confirmed to be highly methylated in tumors and unmethylated in normal mucosal and salivary rinse samples demonstrating cancer-specific methylation in HNSCC tissues. TXNIP and TUSC2 were partially methylated in tumors and normal salivary rinses but unmethylated in normal mucosa. We concluded that GNG7 is a highly specific promoter methylated gene associated with HNSCC. In addition, TXNIP and TUSC2 are also potential biomarkers for HNSCC.
Collapse
Affiliation(s)
- Semra Demokan
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wang D, Zhang Y, Huang Y, Li P, Wang M, Wu R, Cheng L, Zhang W, Zhang Y, Li B, Wang C, Guo Z. Comparison of different normalization assumptions for analyses of DNA methylation data from the cancer genome. Gene 2012; 506:36-42. [PMID: 22771920 DOI: 10.1016/j.gene.2012.06.075] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 06/21/2012] [Accepted: 06/22/2012] [Indexed: 01/02/2023]
Abstract
Nowadays, some researchers normalized DNA methylation arrays data in order to remove the technical artifacts introduced by experimental differences in sample preparation, array processing and other factors. However, other researchers analyzed DNA methylation arrays without performing data normalization considering that current normalizations for methylation data may distort real differences between normal and cancer samples because cancer genomes may be extensively subject to hypomethylation and the total amount of CpG methylation might differ substantially among samples. In this study, using eight datasets by Infinium HumanMethylation27 assay, we systemically analyzed the global distribution of DNA methylation changes in cancer compared to normal control and its effect on data normalization for selecting differentially methylated (DM) genes. We showed more differentially methylated (DM) genes could be found in the Quantile/Lowess-normalized data than in the non-normalized data. We found the DM genes additionally selected in the Quantile/Lowess-normalized data showed significantly consistent methylation states in another independent dataset for the same cancer, indicating these extra DM genes were effective biological signals related to the disease. These results suggested normalization can increase the power of detecting DM genes in the context of diagnostic markers which were usually characterized by relatively large effect sizes. Besides, we evaluated the reproducibility of DM discoveries for a particular cancer type, and we found most of the DM genes additionally detected in one dataset showed the same methylation directions in the other dataset for the same cancer type, indicating that these DM genes were effective biological signals in the other dataset. Furthermore, we showed that some DM genes detected from different studies for a particular cancer type were significantly reproducible at the functional level.
Collapse
Affiliation(s)
- Dong Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Uzhachenko R, Issaeva N, Boyd K, Ivanov SV, Carbone DP, Ivanova AV. Tumour suppressor Fus1 provides a molecular link between inflammatory response and mitochondrial homeostasis. J Pathol 2012; 227:456-69. [PMID: 22513871 DOI: 10.1002/path.4039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 04/04/2012] [Accepted: 04/07/2012] [Indexed: 02/04/2023]
Abstract
Fus1, encoded by a 3p21.3 tumour suppressor gene, is down-regulated, mutated or lost in the majority of inflammatory thoracic malignancies. The mitochondrial localization of Fus1 stimulated us to investigate how Fus1 modulates inflammatory response and mitochondrial function in a mouse model of asbestos-induced peritoneal inflammation. Asbestos treatment resulted in a decreased Fus1 expression in wild-type (WT) peritoneal immune cells, suggesting that asbestos exposure may compromise the Fus1-mediated inflammatory response. Untreated Fus1(-/-) mice had an ~eight-fold higher proportion of peritoneal granulocytes than Fus1(+/+) mice, pointing at ongoing chronic inflammation. Fus1(-/-) mice exhibited a perturbed inflammatory response to asbestos, reflected in decreased immune organ weight and peritoneal fluid protein concentration, along with an increased proportion of peritoneal macrophages. Fus1(-/-) immune cells showed augmented asbestos-induced activation of key inflammatory, anti-oxidant and genotoxic stress response proteins ERK1/2, NFκB, SOD2, γH2AX, etc. Moreover, Fus1(-/-) mice demonstrated altered dynamics of pro- and anti-inflammatory cytokine expression, such as IFNγ, TNFα, IL-1A, IL-1B and IL-10. 'Late' response cytokine Ccl5 was persistently under-expressed in Fus1(-/-) immune cells at both basal and asbestos-activated states. We observed an asbestos-related difference in the size of CD3(+) CD4(-) CD8(-) DN T cell subset that was expanded four-fold in Fus1(-/-) mice. Finally, we demonstrated Fus1-dependent basal and asbestos-induced changes in major mitochondrial parameters (ROS production, mitochondrial potential and UCP2 expression) in Fus1(-/-) immune cells and in Fus1-depleted cancer cells, thus supporting our hypothesis that Fus1 establishes its immune- and tumour-suppressive activities via regulation of mitochondrial homeostasis.
Collapse
Affiliation(s)
- Roman Uzhachenko
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | |
Collapse
|
28
|
Jean D, Daubriac J, Le Pimpec-Barthes F, Galateau-Salle F, Jaurand MC. Molecular changes in mesothelioma with an impact on prognosis and treatment. Arch Pathol Lab Med 2012; 136:277-93. [PMID: 22372904 DOI: 10.5858/arpa.2011-0215-ra] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT In recent decades, research on malignant pleural mesothelioma (MPM) has been developed to improve patients' outcomes by increasing the level of confidence in MPM diagnosis and prognosis. OBJECTIVE To summarize data on genetic and epigenetic abnormalities in MPM that may be of interest for a better management of patients with MPM. DATA SOURCES Data were obtained from scientific publications on genetic and epigenetic abnormalities in MPM by studying gene mutations, DNA methylation, and gene and microRNA expression profiling. CONCLUSIONS Molecular changes in MPM consist in altered expression and in activation or inactivation of critical genes in oncogenesis, especially tumor suppressor genes at the INK4 and NF2 loci. Activation of membrane receptor tyrosine kinases and deregulation of signaling pathways related to differentiation, survival, proliferation, apoptosis, cell cycle control, metabolism, migration, and invasion have been demonstrated. Alterations that could be targeted at a global level (methylation) have been recently reported. Experimental research has succeeded especially in abolishing proliferation and triggering apoptosis in MPM cells. So far, targeted clinical approaches focusing on receptor tyrosine kinases have had limited success. Molecular analyses of series of MPM cases have shown that defined alterations are present in MPM subsets, consistent with interindividual variations of molecular alterations, and suggesting that identification of patient subgroups will be essential to develop more specific therapies.
Collapse
Affiliation(s)
- Didier Jean
- INSERM, U, Université Paris Descartes, UMR-S, Paris, France
| | | | | | | | | |
Collapse
|
29
|
Van Hee VC, Kaufman JD, Budinger GRS, Mutlu GM. Update in environmental and occupational medicine 2009. Am J Respir Crit Care Med 2010; 181:1174-80. [PMID: 20516491 DOI: 10.1164/rccm.201002-0183up] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Victor C Van Hee
- Occupational and Environmental Medicine Program, Department of Medicine and Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | | | | | | |
Collapse
|