1
|
Pennisi G, Bruzzaniti P, Burattini B, Piaser Guerrato G, Della Pepa GM, Sturiale CL, Lapolla P, Familiari P, La Pira B, D’Andrea G, Olivi A, D’Alessandris QG, Montano N. Advancements in Telomerase-Targeted Therapies for Glioblastoma: A Systematic Review. Int J Mol Sci 2024; 25:8700. [PMID: 39201386 PMCID: PMC11354571 DOI: 10.3390/ijms25168700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Glioblastoma (GBM) is a primary CNS tumor that is highly lethal in adults and has limited treatment options. Despite advancements in understanding the GBM biology, the standard treatment for GBM has remained unchanged for more than a decade. Only 6.8% of patients survive beyond five years. Telomerase, particularly the hTERT promoter mutations present in up to 80% of GBM cases, represents a promising therapeutic target due to its role in sustaining telomere length and cancer cell proliferation. This review examines the biology of telomerase in GBM and explores potential telomerase-targeted therapies. We conducted a systematic review following the PRISMA-P guidelines in the MEDLINE/PubMed and Scopus databases, from January 1995 to April 2024. We searched for suitable articles by utilizing the terms "GBM", "high-grade gliomas", "hTERT" and "telomerase". We incorporated studies addressing telomerase-targeted therapies into GBM studies, excluding non-English articles, reviews, and meta-analyses. We evaluated a total of 777 records and 46 full texts, including 36 studies in the final review. Several compounds aimed at inhibiting hTERT transcription demonstrated promising preclinical outcomes; however, they were unsuccessful in clinical trials owing to intricate regulatory pathways and inadequate pharmacokinetics. Direct hTERT inhibitors encountered numerous obstacles, including a prolonged latency for telomere shortening and the activation of the alternative lengthening of telomeres (ALT). The G-quadruplex DNA stabilizers appeared to be potential indirect inhibitors, but further clinical studies are required. Imetelstat, the only telomerase inhibitor that has undergone clinical trials, has demonstrated efficacy in various cancers, but its efficacy in GBM has been limited. Telomerase-targeted therapies in GBM is challenging due to complex hTERT regulation and inadequate inhibitor pharmacokinetics. Our study demonstrates that, despite promising preclinical results, no Telomerase inhibitors have been approved for GBM, and clinical trials have been largely unsuccessful. Future strategies may include Telomerase-based vaccines and multi-target inhibitors, which may provide more effective treatments when combined with a better understanding of telomere dynamics and tumor biology. These treatments have the potential to be integrated with existing ones and to improve the outcomes for patients with GBM.
Collapse
Affiliation(s)
- Giovanni Pennisi
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
- Department of Neurosurgery, F. Spaziani Hospital, 03100 Frosinone, Italy; (B.L.P.); (G.D.)
| | - Placido Bruzzaniti
- Department of Neurosurgery, F. Spaziani Hospital, 03100 Frosinone, Italy; (B.L.P.); (G.D.)
- Department of Human Neurosciences, Division of Neurosurgery, Policlinico Umberto I University Hospital, Sapienza, University of Rome, 00157 Rome, Italy;
| | - Benedetta Burattini
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | - Giacomo Piaser Guerrato
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | - Giuseppe Maria Della Pepa
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | - Carmelo Lucio Sturiale
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | | | - Pietro Familiari
- Department of Human Neurosciences, Division of Neurosurgery, Policlinico Umberto I University Hospital, Sapienza, University of Rome, 00157 Rome, Italy;
| | - Biagia La Pira
- Department of Neurosurgery, F. Spaziani Hospital, 03100 Frosinone, Italy; (B.L.P.); (G.D.)
| | - Giancarlo D’Andrea
- Department of Neurosurgery, F. Spaziani Hospital, 03100 Frosinone, Italy; (B.L.P.); (G.D.)
| | - Alessandro Olivi
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | - Quintino Giorgio D’Alessandris
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | - Nicola Montano
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| |
Collapse
|
2
|
Kumar N, Sethi G. Telomerase and hallmarks of cancer: An intricate interplay governing cancer cell evolution. Cancer Lett 2023; 578:216459. [PMID: 37863351 DOI: 10.1016/j.canlet.2023.216459] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/02/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
Transformed cells must acquire specific characteristics to be malignant. Weinberg and Hanahan characterize these characteristics as cancer hallmarks. Though these features are independently driven, substantial signaling crosstalk in transformed cells efficiently promotes these feature acquisitions. Telomerase is an enzyme complex that maintains telomere length. However, its main component, Telomere reverse transcriptase (TERT), has been found to interact with various signaling molecules like cMYC, NF-kB, BRG1 and cooperate in transcription and metabolic reprogramming, acting as a strong proponent of malignant features such as cell death resistance, sustained proliferation, angiogenesis activation, and metastasis, among others. It allows cells to avoid replicative senescence and achieve endless replicative potential. This review summarizes both the canonical and noncanonical functions of TERT and discusses how they promote cancer hallmarks. Understanding the role of Telomerase in promoting cancer hallmarks provides vital insight into the underlying mechanism of cancer genesis and progression and telomerase intervention as a possible therapeutic target for cancer treatment. More investigation into the precise molecular mechanisms of telomerase-mediated impacts on cancer hallmarks will contribute to developing more focused and customized cancer treatment methods.
Collapse
Affiliation(s)
- Naveen Kumar
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, 138673, Singapore
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
3
|
Ibrahim IH, Ali OSM, El-Sahar AA, Elrefaei M, El-Sheikh N. Telomerase Expression Related with Poor Immune Response to HCV Core Antigen in Egyptian HCV Patients' PBMCs. J Clin Exp Hepatol 2023; 13:1008-1016. [PMID: 37975051 PMCID: PMC10643511 DOI: 10.1016/j.jceh.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/12/2023] [Indexed: 11/19/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most common primary malignant tumor of the liver. Aim This study aimed to assess serum human telomerase enzyme (hTERT) levels and their relation to the progression of liver disease. Also, it aimed to assess the effect of hepatitis C virus (HCV) core protein on memory T-cells in HCV patients with or without HCC and the correlation between memory cell phenotype and the progression of the disease in the same patients. Methods HTERT level in serum was assessed through relative quantitative RT-PCR. Flow cytometric analysis was used to assess T-cell responsiveness (as IFN- γ secretion) before and after stimulation with HCV core protein and the memory CD8+ cell phenotype using several differentiation markers. Results HTERT was found to be increased in a stepwise manner upon comparing its level in controls, chronic hepatitis patients, cirrhotic patients, and HCC patients. T-cells showed a similar manner of stepwise decrease in response (decreased IFN- γ secretion) in HCC patients compared to HCV patients without HCC and controls. Also, late differentiated memory cells (CD8+, CD27-, CD28-, CD45RA+, and CCR7-) were depleted in HCC patients compared to HCV patients without HCC. Conclusion These results suggest a negative correlation between hTERT and IFN- γ secretion by T-cells in HCV patients and that this relationship, along with the depletion of late differentiated memory cells, could help the progression of liver disease to HCC.
Collapse
Affiliation(s)
- Iman H. Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Egypt
| | - Ola Sayed M. Ali
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Egypt
| | - Adel A. El-Sahar
- Department of Tropical Medicine, Faculty of Medicine (Boys), Al-Azhar University, Egypt
| | - Mohamed Elrefaei
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | - Nabila El-Sheikh
- Molecular Immunology Unit for Infectious Diseases, Department of Microbiology and Immunology, Faculty of Medicine (Girls), Al-Azhar University, Egypt
| |
Collapse
|
4
|
Jia Y, Yao P, Li J, Wei X, Liu X, Wu H, Wang W, Feng C, Li C, Zhang Y, Cai Y, Zhang S, Ma X. Causal associations of Sjögren's syndrome with cancers: a two-sample Mendelian randomization study. Arthritis Res Ther 2023; 25:171. [PMID: 37715206 PMCID: PMC10503000 DOI: 10.1186/s13075-023-03157-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/30/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Several observational studies have explored the associations between Sjögren's syndrome (SS) and certain cancers. Nevertheless, the causal relationships remain unclear. Mendelian randomization (MR) method was used to investigate the causality between SS and different types of cancers. METHODS We conducted the two-sample Mendelian randomization with the public genome-wide association studies (GWASs) summary statistics in European population to evaluate the causality between SS and nine types of cancers. The sample size varies from 1080 to 372,373. The inverse variance weighted (IVW) method was used to estimate the causal effects. A Bonferroni-corrected threshold of P < 0.0031 was considered significant, and P value between 0.0031 and 0.05 was considered to be suggestive of an association. Sensitivity analysis was performed to validate the causality. Moreover, additional analysis was used to assess the associations between SS and well-accepted risk factors of cancers. RESULTS After correcting the heterogeneity and horizontal pleiotropy, the results indicated that patients with SS were significantly associated with an increased risk of lymphomas (odds ratio [OR] = 1.0010, 95% confidence interval [CI]: 1.0005-1.0015, P = 0.0002) and reduced risks of prostate cancer (OR = 0.9972, 95% CI: 0.9960-0.9985, P = 2.45 × 10-5) and endometrial cancer (OR = 0.9414, 95% CI: 0.9158-0.9676, P = 1.65 × 10-5). Suggestive associations were found in liver and bile duct cancer (OR = 0.9999, 95% CI: 0.9997-1.0000, P = 0.0291) and cancer of urinary tract (OR = 0.9996, 95% CI: 0.9992-1.0000, P = 0.0281). No causal effect of SS on other cancer types was detected. Additional MR analysis indicated that causal effects between SS and cancers were not mediated by the well-accepted risk factors of cancers. No evidence of the causal relationship was observed for cancers on SS. CONCLUSIONS SS had significant causal relationships with lymphomas, prostate cancer, and endometrial cancer, and suggestive evidence of association was found in liver and bile duct cancer and cancer of urinary tract, indicating that SS may play a vital role in the incidence of these malignancies.
Collapse
Affiliation(s)
- Yiwei Jia
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Peizhuo Yao
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Jia Li
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Xinyu Wei
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Xuanyu Liu
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Huizi Wu
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Weiwei Wang
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Cong Feng
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Chaofan Li
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Yu Zhang
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Yifan Cai
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Shuqun Zhang
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China.
| | - Xingcong Ma
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China.
| |
Collapse
|
5
|
Li J, Feng Y, Zhao J, Fang Z, Liu H. Telomerase reverse transcriptase promotes angiogenesis in neonatal rats after hypoxic-ischemic brain damage. PeerJ 2022; 10:e14220. [PMID: 36299510 PMCID: PMC9590416 DOI: 10.7717/peerj.14220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/20/2022] [Indexed: 01/24/2023] Open
Abstract
Background Angiogenesis is an endogenous repair mechanism following hypoxic-ischemic brain damage (HIBD). Interestingly, recent studies have shown that angiogenesis can be regulated by telomerase reverse transcriptase (TERT), a critical component of telomerase. As telomerase reverse transcriptase can promote angiogenesis after stroke, we hypothesized that it could also promote angiogenesis after HIBD. To test this hypothesis, we developed in vivo and in vitro HIBD models in neonatal rats. Methods TERT was overexpressed by lentivirus and adenovirus infection, and levels were measured using quantitative real-time polymerase chain reaction. We used a cell counting kit to quantify the proliferation rate of brain microvascular endothelial cells (BMECs), and immunofluorescence staining to measure CD34 expression levels. A microvessel formation assay was used to evaluate angiogenesis. Blood-brain barrier (BBB) integrity was assessed using immunohistochemical staining for ZO-1 and Evans Blue staining. Lastly, the expression level of Notch-1 was measured by western blotting. Results Overexpression of TERT promoted the proliferation of BMECs after hypoxic-ischemic damage in vitro. TERT overexpression increased the formation of microvessels in the neonatal brain after HIBD both in vivo and in vitro. Overexpression of TERT improved BBB integrity in the brains of neonatal rats after HIBD. In addition, the expression level of Notch-1 was increased in BMECs following oxygen glucose deprivation, and overexpression of TERT further increased Notch-1 expression levels in BMECs following oxygen glucose deprivation. Discussion Our results reveal that telomerase reverse transcriptase promotes angiogenesis and maintains the integrity of the blood-brain barrier after neonatal hypoxic-ischemic brain damage. Furthermore, the Notch-1 signaling pathway appears to contribute to the angiogenic function of telomerase reverse transcriptase. This protective effect of telomerase reverse transcriptase opens new horizons for future investigations aimed at uncovering the full potential of telomerase reverse transcriptase as a promising new target for the treatment of hypoxic-ischemic encephalopathy.
Collapse
Affiliation(s)
- Jiao Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Feng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jing Zhao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhi Fang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Haiting Liu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Telomerase in Cancer: Function, Regulation, and Clinical Translation. Cancers (Basel) 2022; 14:cancers14030808. [PMID: 35159075 PMCID: PMC8834434 DOI: 10.3390/cancers14030808] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cells undergoing malignant transformation must circumvent replicative senescence and eventual cell death associated with progressive telomere shortening that occurs through successive cell division. To do so, malignant cells reactivate telomerase to extend their telomeres and achieve cellular immortality, which is a “Hallmark of Cancer”. Here we review the telomere-dependent and -independent functions of telomerase in cancer, as well as its potential as a biomarker and therapeutic target to diagnose and treat cancer patients. Abstract During the process of malignant transformation, cells undergo a series of genetic, epigenetic, and phenotypic alterations, including the acquisition and propagation of genomic aberrations that impart survival and proliferative advantages. These changes are mediated in part by the induction of replicative immortality that is accompanied by active telomere elongation. Indeed, telomeres undergo dynamic changes to their lengths and higher-order structures throughout tumor formation and progression, processes overseen in most cancers by telomerase. Telomerase is a multimeric enzyme whose function is exquisitely regulated through diverse transcriptional, post-transcriptional, and post-translational mechanisms to facilitate telomere extension. In turn, telomerase function depends not only on its core components, but also on a suite of binding partners, transcription factors, and intra- and extracellular signaling effectors. Additionally, telomerase exhibits telomere-independent regulation of cancer cell growth by participating directly in cellular metabolism, signal transduction, and the regulation of gene expression in ways that are critical for tumorigenesis. In this review, we summarize the complex mechanisms underlying telomere maintenance, with a particular focus on both the telomeric and extratelomeric functions of telomerase. We also explore the clinical utility of telomeres and telomerase in the diagnosis, prognosis, and development of targeted therapies for primary, metastatic, and recurrent cancers.
Collapse
|
7
|
Gu H, Zhang Y, Zeng W, Xia Y. Participation of interferons in psoriatic inflammation. Cytokine Growth Factor Rev 2021; 64:12-20. [PMID: 34953718 DOI: 10.1016/j.cytogfr.2021.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 12/17/2022]
Abstract
Interferons are multifunctional cytokines not expressed in the skin under normal physiological conditions. However, they are overexpressed in serum and skin lesions of patients with psoriasis and play an important role in the pathogenesis of the disease. Interferons act directly on skin resident cells and recruit and modulate inflammatory cells, thereby exacerbating psoriatic inflammation. They upregulate the expression of relevant cytokines and chemokines, facilitate excessive proliferation of keratinocytes, and enhance the formation of poorly differentiated dermal microvessels. In this review, we summarized the pathogenic effect of interferons on psoriasis and also discussed the therapeutic strategies targeting interferons.
Collapse
Affiliation(s)
- Hanjiang Gu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yufei Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weihui Zeng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
8
|
Xu LQ, Yao LJ, Jiang D, Zhou LJ, Chen M, Liao WZ, Zou WH, Peng HJ. A uracil auxotroph Toxoplasma gondii exerting immunomodulation to inhibit breast cancer growth and metastasis. Parasit Vectors 2021; 14:601. [PMID: 34895326 PMCID: PMC8665513 DOI: 10.1186/s13071-021-05032-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Breast cancer is the most common cause of cancer-related death among women, and prognosis is especially poor for patients with triple-negative breast cancer (TNBC); therefore, there is an urgent need for new effective therapies. Recent studies have demonstrated that the uracil auxotroph Toxoplasma gondii vaccine displays anti-tumor effects. Here, we examined the immunotherapy effects of an attenuated uracil auxotroph strain of T. gondii against 4T1 murine breast cancer. METHODS We constructed a uracil auxotroph T. gondii RH strain via orotidine 5'-monophosphate decarboxylase gene deletion (RH-Δompdc) with CRISPR/Cas9 technology. The strain's virulence in the T. gondii-infected mice was determined in vitro and in vivo by parasite replication assay, plaque assay, parasite burden detection in mice peritoneal fluids and survival analysis. The immunomodulation ability of the strain was evaluated by cytokine detection. Its anti-tumor effect was evaluated after its in situ inoculation into 4T1 tumors in a mouse model; the tumor volume was measured, and the 4T1 lung metastasis was detected by hematoxylin and eosin and Ki67 antibody staining, and the cytokine levels were measured by an enzyme-linked immunosorbent assay. RESULTS The RH-Δompdc strain proliferated normally when supplemented with uracil, but it was unable to propagate without the addition of uracil and in vivo, which suggested that it was avirulent to the hosts. This mutant showed vaccine characteristics that could induce intense immune responses both in vitro and in vivo by significantly boosting the expression of inflammatory cytokines. Inoculation of RH-Δompdc in situ into the 4T1 tumor inhibited tumor growth, reduced lung metastasis, promoted the survival of the tumor-bearing mice and increased the secretion of Th1 cytokines, including interleukin-12 (IL-12) and interferon-γ (INF-δ), in both the serum and tumor microenvironment (TME). CONCLUSION Inoculation of the uracil auxotroph RH-Δompdc directly into the 4T1 tumor stimulated anti-infection and anti-tumor immunity in mice, and resulted in inhibition of tumor growth and metastasis, promotion of the survival of the tumor-bearing mice and increased secretion of IL-12 and IFN-γ in both the serum and TME. Our findings suggest that the immunomodulation caused by RH-Δompdc could be a potential anti-tumor strategy.
Collapse
Affiliation(s)
- Li-Qing Xu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Li-Jie Yao
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Dan Jiang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Li-Juan Zhou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Min Chen
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Wen-Zhong Liao
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Wei-Hao Zou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Hong-Juan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China.
| |
Collapse
|
9
|
Parulekar A, Choksi A, Taye N, Totakura KVS, Firmal P, Kundu GC, Chattopadhyay S. SMAR1 suppresses the cancer stem cell population via hTERT repression in colorectal cancer cells. Int J Biochem Cell Biol 2021; 141:106085. [PMID: 34551340 DOI: 10.1016/j.biocel.2021.106085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 09/09/2021] [Accepted: 09/16/2021] [Indexed: 11/25/2022]
Abstract
One of the hallmarks of a cancer cell is the ability for indefinite proliferation leading to the immortalization of the cell. Activation of several signaling pathways leads to the immortalization of cancer cells via the reactivation of enzyme telomerase (hTERT). hTERT is active in germ cells, stem cells and also cancer cells. An earlier report from our lab suggests that SMAR1, a tumor suppressor protein, is significantly downregulated in the higher grades of colorectal cancers. Our study identifies SMAR1 as a transcriptional repressor of hTERT. We find that SMAR1 interacts with HDAC1/mSin3a co-repressor complex at the hTERT promoter and brings about HDAC1-mediated transcriptional repression of the promoter. Most solid tumors including colorectal cancer reactivate hTERT expression as it confers several advantages to the cancer cells like increased proliferation and angiogenesis. One of these non-canonical functions of hTERT is inducing the pool of cancer stem cell population. We find that in the CD133HighCD44High cancer stem cells population, SMAR1 expression is highly diminished leading to elevated hTERT expression. We also find that knockdown of SMAR1 promotes total CD133+CD44+ population and impart enhanced sphere-forming ability to the colorectal cancer cells. SMAR1 also inhibits invasion and metastasis in colorectal cancer cell lines via repression of hTERT. Our study provides evidence that downregulation of SMAR1 causes activation of hTERT leading to an increase in the cancer stem cell phenotype in colorectal cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Gopal C Kundu
- National Centre for Cell Science, Pune, India; Kalinga Institute of Industrial Technology, Bhubaneswar, India
| | - Samit Chattopadhyay
- National Centre for Cell Science, Pune, India; Birla Institute of Technology and Science, Goa, India.
| |
Collapse
|
10
|
Applications of CRISPR-Cas9 Technology to Genome Editing in Glioblastoma Multiforme. Cells 2021; 10:cells10092342. [PMID: 34571991 PMCID: PMC8468137 DOI: 10.3390/cells10092342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive malignancy of the brain and spinal cord with a poor life expectancy. The low survivability of GBM patients can be attributed, in part, to its heterogeneity and the presence of multiple genetic alterations causing rapid tumor growth and resistance to conventional therapy. The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-CRISPR associated (Cas) nuclease 9 (CRISPR-Cas9) system is a cost-effective and reliable gene editing technology, which is widely used in cancer research. It leads to novel discoveries of various oncogenes that regulate autophagy, angiogenesis, and invasion and play important role in pathogenesis of various malignancies, including GBM. In this review article, we first describe the principle and methods of delivery of CRISPR-Cas9 genome editing. Second, we summarize the current knowledge and major applications of CRISPR-Cas9 to identifying and modifying the genetic regulators of the hallmark of GBM. Lastly, we elucidate the major limitations of current CRISPR-Cas9 technology in the GBM field and the future perspectives. CRISPR-Cas9 genome editing aids in identifying novel coding and non-coding transcriptional regulators of the hallmarks of GBM particularly in vitro, while work using in vivo systems requires further investigation.
Collapse
|
11
|
Zhang X, Wang S, Zhu Y, Zhang M, Zhao Y, Yan Z, Wang Q, Li X. Double-edged effects of interferons on the regulation of cancer-immunity cycle. Oncoimmunology 2021; 10:1929005. [PMID: 34262796 PMCID: PMC8253121 DOI: 10.1080/2162402x.2021.1929005] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Interferons (IFNs) are a large family of pleiotropic cytokines that regulate both innate and adaptive immunity and show anti-cancer effects in various cancer types. Moreover, it was revealed that IFN signaling plays critical roles in the success of cancer therapy strategies, thereby enhancing their therapeutic effects. However, IFNs have minimal or even adverse effects on cancer eradication, and mediate cancer immune escape in some instances. Thus, IFNs have a double-edged effect on the cancer immune response. Recent studies suggest that IFNs regulate each step of the cancer immunity-cycle, consisting of cancer antigen release, presentation of antigens and activation of T cells, trafficking and infiltration of effector T cells into the tumor microenvironment, and recognition and killing of cancer cells, which contributes to our understanding of the mechanisms of IFNs in regulating cancer immunity. In this review, we focus on IFNs and cancer immunity and elaborate on the roles of IFNs in regulating the cancer-immunity cycle.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Stomatology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Department of Pathology, Harbin Medical University, Harbin, China
| | - Song Wang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yuanyuan Zhu
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Minghui Zhang
- Department of Oncology, Chifeng City Hospital, Chifeng, China
| | - Yan Zhao
- Department of Oncology, Chifeng City Hospital, Chifeng, China
| | - Zhengbin Yan
- Department of Stomatology, the PeopIe's Hospital of Longhua, Shenzhen, China
| | - Qiuxu Wang
- Department of Stomatology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Department of Stomatology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Xiaobo Li
- Department of Stomatology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Department of Pathology, Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
Herbener VJ, Burster T, Goreth A, Pruss M, von Bandemer H, Baisch T, Fitzel R, Siegelin MD, Karpel-Massler G, Debatin KM, Westhoff MA, Strobel H. Considering the Experimental use of Temozolomide in Glioblastoma Research. Biomedicines 2020; 8:E151. [PMID: 32512726 PMCID: PMC7344626 DOI: 10.3390/biomedicines8060151] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/30/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022] Open
Abstract
Temozolomide (TMZ) currently remains the only chemotherapeutic component in the approved treatment scheme for Glioblastoma (GB), the most common primary brain tumour with a dismal patient's survival prognosis of only ~15 months. While frequently described as an alkylating agent that causes DNA damage and thus-ultimately-cell death, a recent debate has been initiated to re-evaluate the therapeutic role of TMZ in GB. Here, we discuss the experimental use of TMZ and highlight how it differs from its clinical role. Four areas could be identified in which the experimental data is particularly limited in its translational potential: 1. transferring clinical dosing and scheduling to an experimental system and vice versa; 2. the different use of (non-inert) solvent in clinic and laboratory; 3. the limitations of established GB cell lines which only poorly mimic GB tumours; and 4. the limitations of animal models lacking an immune response. Discussing these limitations in a broader biomedical context, we offer suggestions as to how to improve transferability of data. Finally, we highlight an underexplored function of TMZ in modulating the immune system, as an example of where the aforementioned limitations impede the progression of our knowledge.
Collapse
Affiliation(s)
- Verena J. Herbener
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Alicia Goreth
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Maximilian Pruss
- Department of Gynecology and Obstetrics, Medical Faculty, University Hospital of the Heinrich-Heine-University Duesseldorf, D-40225 Duesseldorf, Germany;
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Hélène von Bandemer
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Tim Baisch
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Rahel Fitzel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Markus D. Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Georg Karpel-Massler
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| |
Collapse
|
13
|
Liu N, Guo XH, Liu JP, Cong YS. Role of telomerase in the tumour microenvironment. Clin Exp Pharmacol Physiol 2019; 47:357-364. [PMID: 31799699 DOI: 10.1111/1440-1681.13223] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/28/2019] [Accepted: 12/01/2019] [Indexed: 12/14/2022]
Abstract
Telomeres are specialized genomic structures that protect chromosomal ends to maintain genomic stability. Telomeric length is primarily regulated by the telomerase complex, essentially consisting of an RNA template (TERC), an enzymatic subunit (telomerase reverse transcriptase, TERT). In humans, telomerase activity is repressed during embryonic differentiation and is absent in most somatic cells. However, it is upregulated or reactivated in 80%-90% of the primary tumours in humans. The human TERT (hTERT) plays a pivotal role in cellular immortality and tumourigenesis. However, the molecular mechanisms of telomerase functioning in cancer have not been fully understood beyond the telomere maintenance. Several research groups, including ours, have demonstrated that hTERT possesses vital functions independent of its telomere maintenance, including angiogenesis, inflammation, cancer cell stemness, and epithelial-mesenchymal transformation (EMT). All these telomere-independent activities of hTERT may contribute to the regulation of the dynamics and homeostasis of the tumour microenvironment (TME), thereby promoting tumour growth and development. Cancer progression and metastasis largely depend upon the interactions between cancer cells and their microenvironment. In this review, the involvement of TERT in the tumour microenvironment and the underlying implications in cancer therapeutics have been summarized.
Collapse
Affiliation(s)
- Ning Liu
- College of Food Sciences and Technology, Shanghai Ocean University, Shanghai, China
| | - Xue-Hua Guo
- College of Food Sciences and Technology, Shanghai Ocean University, Shanghai, China
| | - Jun-Ping Liu
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Yu-Sheng Cong
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, China.,Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou, China
| |
Collapse
|
14
|
Improved Antitumor Efficacy of Combined Vaccine Based on the Induced HUVECs and DC-CT26 Against Colorectal Carcinoma. Cells 2019; 8:cells8050494. [PMID: 31121964 PMCID: PMC6562839 DOI: 10.3390/cells8050494] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 12/24/2022] Open
Abstract
Angiogenesis is essential for the development, growth, and metastasis of solid tumors. Vaccination with viable human umbilical vein endothelial cells (HUVECs) has been used for antitumor angiogenesis. However, the limited immune response induced by HUVECs hinders their clinical application. In the present study, we found that HUVECs induced by a tumor microenvironment using the supernatant of murine CT26 colorectal cancer cells exerted a better antiangiogenic effect than HUVECs themselves. The inhibitory effect on tumor growth in the induced HUVEC group was significantly better than that of the HUVEC group, and the induced HUVEC group showed a strong inhibition in CD31-positive microvessel density in the tumor tissues. Moreover, the level of anti-induced HUVEC membrane protein antibody in mouse serum was profoundly higher in the induced HUVEC group than in the HUVEC group. Based on this, the antitumor effect of a vaccine with a combination of induced HUVECs and dendritic cell-loading CT26 antigen (DC-CT26) was evaluated. Notably, the microvessel density of tumor specimens was significantly lower in the combined vaccine group than in the control groups. Furthermore, the spleen index, the killing effect of cytotoxic T lymphocytes (CTLs), and the concentration of interferon-γ in the serum were enhanced in the combined vaccine group. Based on these results, the combined vaccine targeting both tumor angiogenesis and tumor cells may be an attractive and effective cancer immunotherapy strategy.
Collapse
|
15
|
Romaniuk A, Paszel-Jaworska A, Totoń E, Lisiak N, Hołysz H, Królak A, Grodecka-Gazdecka S, Rubiś B. The non-canonical functions of telomerase: to turn off or not to turn off. Mol Biol Rep 2018; 46:1401-1411. [PMID: 30448892 DOI: 10.1007/s11033-018-4496-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022]
Abstract
Telomerase is perceived as an immortality enzyme that enables passing the Hayflick limit. Its main function is telomere restoration but only in a limited group of cells, including cancer cells. Since it is found in a vast majority of cancer cells, it became a natural target for cancer therapy. However, it has much more functions than just altering the metabolism of telomeres-it also reveals numerous so-called non-canonical functions. Thus, a question arises whether it is always beneficial to turn it off when planning a cancer strategy and considering potential side effects? The purpose of this review is to discuss some of the recent discoveries about telomere-independent functions of telomerase in the context of cancer therapy and potential side effects.
Collapse
Affiliation(s)
- Aleksandra Romaniuk
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | - Anna Paszel-Jaworska
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | - Ewa Totoń
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | - Natalia Lisiak
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | - Hanna Hołysz
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | - Anna Królak
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | | | - Błażej Rubiś
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland.
| |
Collapse
|
16
|
Li HL, Song J, Yong HM, Hou PF, Chen YS, Song WB, Bai J, Zheng JN. PinX1: structure, regulation and its functions in cancer. Oncotarget 2018; 7:66267-66275. [PMID: 27556185 PMCID: PMC5323232 DOI: 10.18632/oncotarget.11411] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/09/2016] [Indexed: 02/07/2023] Open
Abstract
PIN2/TRF1-interacting telomerase inhibitor 1 (PinX1) is a novel cloned gene located at human chromosome 8p23, playing a vital role in maintaining telomeres length and chromosome stability. It has been demonstrated to be involved in tumor genesis and progression in most malignancies. However, some researches showed opposing molecular status of PinX1 gene and its expression patterns in several other types of tumors. The pathogenic mechanism of PinX1 expression in human malignancy is not yet clear. Moreover, emerging evidence suggest that PinX1 (especially its TID domain) might be a potential new target cancer treatment. Therefore, PinX1 may be a new potential diagnostic biomarker and therapeutic target for human cancers, and may play different roles in different human cancers. The functions and the mechanisms of PinX1 in various human cancers remain unclear, suggesting the necessity of further extensive works of its role in tumor genesis and progression.
Collapse
Affiliation(s)
- Hai-Long Li
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China.,Department of Urology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jun Song
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu, China.,Department of General Surgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Hong-Mei Yong
- Department of Medical Oncology, Huai'an Hospital to Xuzhou Medical College, Huai'an, Jiangsu, China
| | - Ping-Fu Hou
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Yan-Su Chen
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Wen-Bo Song
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jin Bai
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jun-Nian Zheng
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu, China
| |
Collapse
|
17
|
Pestana A, Vinagre J, Sobrinho-Simões M, Soares P. TERT biology and function in cancer: beyond immortalisation. J Mol Endocrinol 2017; 58:R129-R146. [PMID: 28057768 DOI: 10.1530/jme-16-0195] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/05/2017] [Indexed: 12/23/2022]
Abstract
Evasion of replicative senescence and proliferation without restriction, sometimes designated as immortalisation, is one of the hallmarks of cancer that may be attained through reactivation of telomerase in somatic cells. In contrast to most normal cells in which there is lack of telomerase activity, upregulation of TERT transcription/activity is detected in 80-90% of malignant tumours. In several types of cancer, there is a relationship between the presence of TERT promoter mutations, TERT mRNA expression and clinicopathological features, but the biological bridge between the occurrence of TERT promoter mutations and the aggressive/invasive features displayed by the tumours remains unidentified. We and others have associated the presence of TERT promoter mutations with metastisation/survival in several types of cancer. In follicular cell-derived thyroid cancer, such mutations are associated with worse prognostic features (age of patients, tumour size and tumour stage) as well as with distant metastases, worse response to treatment and poorer survival. In this review, we analyse the data reported in several studies that imply TERT transcription reactivation/activity with cell proliferation, tumour invasion and metastisation. A particular attention is given to the putative connections between TERT transcriptional reactivation and signalling pathways frequently altered in cancer, such as c-MYC, NF-κB and B-Catenin.
Collapse
Affiliation(s)
- Ana Pestana
- Institute of Molecular Pathology and ImmunologyUniversity of Porto (IPATIMUP), Porto, Portugal
- Institute for Research and Innovation in Health (I3S)University of Porto, Porto, Portugal
| | - João Vinagre
- Institute of Molecular Pathology and ImmunologyUniversity of Porto (IPATIMUP), Porto, Portugal
- Institute for Research and Innovation in Health (I3S)University of Porto, Porto, Portugal
| | - Manuel Sobrinho-Simões
- Institute of Molecular Pathology and ImmunologyUniversity of Porto (IPATIMUP), Porto, Portugal
- Institute for Research and Innovation in Health (I3S)University of Porto, Porto, Portugal
- Medical FacultyUniversity of Porto, Porto, Portugal
- Department of PathologyCentro Hospitalar S. João, Porto, Portugal
- Department of PathologyMedical Faculty, University of Porto, Porto, Portugal
| | - Paula Soares
- Institute of Molecular Pathology and ImmunologyUniversity of Porto (IPATIMUP), Porto, Portugal
- Institute for Research and Innovation in Health (I3S)University of Porto, Porto, Portugal
- Medical FacultyUniversity of Porto, Porto, Portugal
- Department of PathologyMedical Faculty, University of Porto, Porto, Portugal
| |
Collapse
|
18
|
Ghildiyal R, Sen E. CK2 induced RIG-I drives metabolic adaptations in IFNγ-treated glioma cells. Cytokine 2017; 89:219-228. [PMID: 26631910 DOI: 10.1016/j.cyto.2015.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 10/06/2015] [Accepted: 10/20/2015] [Indexed: 12/16/2022]
Abstract
Given the known anti-tumorigenic properties of IFNγ, its effect on glioma cell survival was investigated. Though IFNγ had no effect on glioma cell viability, it induced cell cycle arrest. This was accompanied by increased expression of p53 and retinoic acid inducible gene (RIG-I). While RIG-I had no effect on glioma cell survival, it increased expression of p53 and its downstream target TP53 induced glycolysis and apoptosis regulator (TIGAR). IFNγ induced mitochondrial co-localization of RIG-I was concomitant with its ability to regulate ROS generation, oxidative phosphorylation (OXPHOS) and key enzymes involved in glycolysis and pentose phosphate pathway. Importantly, metabolic gene profiling indicated a suppressed glycolytic pathway in glioma cells upon IFNγ treatment. In addition, IFNγ mediated increase in casein kinase 2 (CK2) expression positively regulated RIG-I expression. These findings demonstrate how IFNγ induced CK2 regulates RIG-I to drive a complex program of metabolic adaptation and redox homeostasis, crucial for determining glioma cell fate.
Collapse
Affiliation(s)
- Ruchi Ghildiyal
- National Brain Research Centre, Manesar 122 051, Haryana, India
| | - Ellora Sen
- National Brain Research Centre, Manesar 122 051, Haryana, India.
| |
Collapse
|
19
|
RNA interference mediated downregulation of human telomerase reverse transcriptase (hTERT) in LN18 cells. Cytotechnology 2016; 68:2311-2321. [PMID: 27757712 DOI: 10.1007/s10616-016-0025-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 08/24/2016] [Indexed: 10/20/2022] Open
Abstract
Human telomerase reverse transcriptase (hTERT) gene is a biomarker for the targeted therapy in various cancers. Presence of increased telomerase activity is a common feature of all cancers including glioblastoma. Both RNA and catalytic subunits of hTERT are the target sites for blocking its activity. The current study focuses on the expression of hTERT in glioblastoma and its regulation using two different novel siRNAs (small interfering RNA). Our patient data demonstrated increased expression of hTERT, which could be correlated with carcinogenesis in glioma. In vitro studies in siRNA transfected LN18 cells confirmed significant cell death (p < 0.05) as evidenced by MTT and trypan blue exclusion assay. These results were further supported by flow cytometry data, which showed significant increase in early and late apoptosis. The hTERT mRNA expression was effectively downregulated by 45 and 39 % with siRNA1 and siRNA2, respectively. These results were further confirmed by immunoblotting analysis (p < 0.05). Our results suggest that both the siRNAs effectively down regulated the expression of hTERT at mRNA and protein levels, thereby decreasing cell viability and proliferation rate. Hence siRNA mediated downregulation of hTERT could be a potential therapeutic avenue in glioblastoma.
Collapse
|
20
|
Liu N, Ding D, Hao W, Yang F, Wu X, Wang M, Xu X, Ju Z, Liu JP, Song Z, Shay JW, Guo Y, Cong YS. hTERT promotes tumor angiogenesis by activating VEGF via interactions with the Sp1 transcription factor. Nucleic Acids Res 2016; 44:8693-8703. [PMID: 27325744 PMCID: PMC5062966 DOI: 10.1093/nar/gkw549] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 06/04/2016] [Accepted: 06/07/2016] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is recognized as an important hallmark of cancer. Although telomerase is thought to be involved in tumor angiogenesis, the evidence and underlying mechanism remain elusive. Here, we demonstrate that human telomerase reverse transcriptase (hTERT) activates vascular epithelial growth factor (VEGF) gene expression through interactions with the VEGF promoter and the transcription factor Sp1. hTERT binds to Sp1 in vitro and in vivo and stimulates angiogenesis in a manner dependent on Sp1. Deletion of the mTert gene in the first generation of Tert null mice compromised tumor growth, with reduced VEGF expression. In addition, we show that hTERT expression levels are positively correlated with those of VEGF in human gastric tumor samples. Together, our results demonstrate that hTERT facilitates tumor angiogenesis by up-regulating VEGF expression through direct interactions with the VEGF gene and the Sp1 transcription factor. These results provide novel insights into hTERT function in tumor progression in addition to its role in telomere maintenance.
Collapse
Affiliation(s)
- Ning Liu
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China
| | - Deqiang Ding
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China
| | - Wanyu Hao
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China
| | - Fan Yang
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China
| | - Xiaoying Wu
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China
| | - Miao Wang
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China
| | - Xiaoling Xu
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China
| | - Zhenyu Ju
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China
| | - Jun-Ping Liu
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China
| | - Zhangfa Song
- Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou 310016, China
| | - Jerry W Shay
- University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yunliang Guo
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yu-Sheng Cong
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China
| |
Collapse
|
21
|
Kursunel MA, Esendagli G. The untold story of IFN-γ in cancer biology. Cytokine Growth Factor Rev 2016; 31:73-81. [DOI: 10.1016/j.cytogfr.2016.07.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/07/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022]
|
22
|
Nijaguna MB, Patil V, Hegde AS, Chandramouli BA, Arivazhagan A, Santosh V, Somasundaram K. An Eighteen Serum Cytokine Signature for Discriminating Glioma from Normal Healthy Individuals. PLoS One 2015; 10:e0137524. [PMID: 26390214 PMCID: PMC4577083 DOI: 10.1371/journal.pone.0137524] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/18/2015] [Indexed: 02/04/2023] Open
Abstract
Glioblastomas (GBM) are largely incurable as they diffusely infiltrate adjacent brain tissues and are difficult to diagnose at early stages. Biomarkers derived from serum, which can be obtained by minimally invasive procedures, may help in early diagnosis, prognosis and treatment monitoring. To develop a serum cytokine signature, we profiled 48 cytokines in sera derived from normal healthy individuals (n = 26) and different grades of glioma patients (n = 194). We divided the normal and grade IV glioma/GBM serum samples randomly into equal sized training and test sets. In the training set, the Prediction Analysis for Microarrays (PAM) identified a panel of 18 cytokines that could discriminate GBM sera from normal sera with maximum accuracy (95.40%) and minimum error (4.60%). The 18-cytokine signature obtained in the training set discriminated GBM sera from normal sera in the test set as well (accuracy 96.55%; error 3.45%). Interestingly, the 18-cytokine signature also differentiated grade II/Diffuse Astrocytoma (DA) and grade III/Anaplastic Astrocytoma (AA) sera from normal sera very efficiently (DA vs. normal–accuracy 96.00%, error 4.00%; AA vs. normal–accuracy 95.83%, error 4.17%). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis using 18 cytokines resulted in the enrichment of two pathways, cytokine-cytokine receptor interaction and JAK-STAT pathways with high significance. Thus our study identified an 18-cytokine signature for distinguishing glioma sera from normal healthy individual sera and also demonstrated the importance of their differential abundance in glioma biology.
Collapse
Affiliation(s)
- Mamatha B. Nijaguna
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Vikas Patil
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Alangar S. Hegde
- Department of Neurosurgery, Sri Satya Sai Institute of Higher Medical Sciences, Bangalore 560066, India
| | - Bangalore A. Chandramouli
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bangalore 560029, India
| | - Arimappamagan Arivazhagan
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bangalore 560029, India
| | - Vani Santosh
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bangalore 560029, India
| | - Kumaravel Somasundaram
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
- * E-mail:
| |
Collapse
|
23
|
de Laurentiis A, Hiscott J, Alcalay M. The TEL-AML1 fusion protein of acute lymphoblastic leukemia modulates IRF3 activity during early B-cell differentiation. Oncogene 2015; 34:6018-28. [DOI: 10.1038/onc.2015.50] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 12/25/2022]
|
24
|
Telomerase: central regulator of all of the hallmarks of cancer. Trends Biochem Sci 2013; 38:426-34. [PMID: 23932019 DOI: 10.1016/j.tibs.2013.07.001] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/26/2013] [Accepted: 07/01/2013] [Indexed: 12/13/2022]
Abstract
The hallmarks of cancer described by Hanahan and Weinberg are properties that cancer cells must possess for successful transformation. It is believed that each of these hallmarks is independently driven. Although elongation of telomeres is thought to be the prime function of reactivated telomerase reverse transcriptase, this activity does not account for all its effects, such as increasing cell proliferation, resistance to apoptosis, and invasion. Recent studies suggest that the telomerase subunit telomerase reverse transcriptase (TERT) has novel molecular functions including transcriptional regulation and metabolic reprogramming. We summarize these functions and discuss how they could directly regulate the various hallmarks of cancer. Finally, we suggest that therapeutics targeting noncanonical telomerase functions may work better than those that target its role in telomere extension.
Collapse
|
25
|
IFN-γ selectively exerts pro-apoptotic effects on tumor-initiating label-retaining colon cancer cells. Cancer Lett 2013; 336:174-84. [DOI: 10.1016/j.canlet.2013.04.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 04/01/2013] [Accepted: 04/26/2013] [Indexed: 12/29/2022]
|
26
|
Chakrabarti M, Banik NL, Ray SK. miR-138 overexpression is more powerful than hTERT knockdown to potentiate apigenin for apoptosis in neuroblastoma in vitro and in vivo. Exp Cell Res 2013; 319:1575-85. [PMID: 23562653 DOI: 10.1016/j.yexcr.2013.02.025] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/21/2013] [Accepted: 02/22/2013] [Indexed: 01/15/2023]
Abstract
Decrease in expression of the tumor suppressor microRNA-138 (miR-138) correlates well with an increase in telomerase activity in many human cancers. The ability of almost all human cancer cells to grow indefinitely is dependent on presence of telomerase activity. The catalytic component of human telomerase reverse transcriptase (hTERT) regulates telomerase activity in most of the human cancers including malignant neuroblastoma. We observed an indirect increase in the expression of miR-138 after the transfection with hTERT short hairpin RNA (shRNA) plasmid in human malignant neuroblastoma SK-N-DZ and SK-N-BE2 cell lines. Transfection with hTERT shRNA plasmid followed by treatment with the flavonoid apigenin (APG) further increased expression of miR-138. Direct transfection with miR-138 mimic was more powerful than transfection with hTERT shRNA plasmid in potentiating efficacy of APG for decreasing cell viability and colony formation capability of both cell lines. Upregulation of miR-138 was also more effective than down regulation of hTERT in enhancing efficacy of APG for induction of apoptosis in malignant neuroblastoma cells in vitro and in vivo. We delineated that apoptosis occurred with induction of molecular components of the extrinsic and intrinsic pathways in SK-N-DZ and SK-N-BE2 cells both in vitro and in vivo. In conclusion, these results demonstrate that direct miR-138 overexpression is more powerful than hTERT down regulation in enhancing pro-apoptotic effect of APG for controlling growth of human malignant neuroblastoma in cell culture and animal models.
Collapse
Affiliation(s)
- Mrinmay Chakrabarti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | | | | |
Collapse
|
27
|
Sequential hTERT knockdown and apigenin treatment inhibited invasion and proliferation and induced apoptosis in human malignant neuroblastoma SK-N-DZ and SK-N-BE2 cells. J Mol Neurosci 2013; 51:187-98. [PMID: 23417743 DOI: 10.1007/s12031-013-9975-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 01/29/2013] [Indexed: 12/23/2022]
Abstract
Human telomerase reverse transcriptase (hTERT) plays a key role in conferring immortality to human malignant neuroblastomas. We first determined differential expression of hTERT in four human malignant neuroblastoma SH-SY5Y, SK-N-DZ, SK-N-BE2, and IMR-32 cell lines. We then used SK-N-DZ and SK-N-BE2 cell lines, which showed the highest expression of hTERT, to investigate the therapeutic effects of sequential hTERT knockdown and apigenin (APG) treatment. We performed cell invasion assay and studied alterations in expression of matrix metalloproteinases and cell cycle regulatory molecules after this combination therapy. We also investigated induction of apoptosis by using in situ Wright staining, Annexin V staining, and Western blotting. Sequential hTERT knockdown and APG treatment significantly downregulated expression of hTERT so as to cause over 90 % inhibition of cell invasion and 70 % induction of apoptosis in both SK-N-DZ and SK-N-BE2 cell lines. Western blotting demonstrated downregulation of the molecules involved in cell invasion and proliferation, but upregulation of the cell cycle inhibitor and apoptosis-inducing molecules. In conclusion, our current results clearly showed that sequential hTERT knockdown and APG treatment could be a promising therapeutic strategy for effective inhibition of invasion and proliferation and induction of apoptosis in hTERT overexpressing malignant neuroblastoma cells.
Collapse
|
28
|
Hsp90 Inhibitors NVP-AUY922 and NVP-BEP800 May Exert a Significant Radiosensitization on Tumor Cells along with a Cell Type-Specific Cytotoxicity. Transl Oncol 2012; 5:356-69. [PMID: 23066444 DOI: 10.1593/tlo.12211] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 06/28/2012] [Accepted: 07/04/2012] [Indexed: 02/04/2023] Open
Abstract
Targeting heat shock protein 90 (Hsp90) provides a promising therapeutic approach to enhance the sensitivity of tumor cells to ionizing radiation (IR). To explore the impact of scheduling drug-IR administration, in the present study, we analyzed the response of lung carcinoma A549 and glioblastoma SNB19 cells to simultaneous drug-IR treatment followed by a long-term drug administration. Cellular response was evaluated at different time intervals after IR-alone, drug-alone, or combined drug-IR treatments by colony counts and expression profiles of Hsp90 and its clients, along with several apoptotic markers and cell cycle-related proteins, as well as by IR-drug-induced cell cycle arrest, DNA damage, and repair. A short 30-minute exposure to either Hsp90 inhibitor did not affect the radiosensitivity of both tumor cell lines. Increasing the duration of post-IR-drug treatment progressively enhanced the sensitivity of SNB19 cells to IR. In contrast, the response of A549 cells to drug-IR combination was largely determined by the cytotoxic effects of both drugs without radiosensitization. Combined drug-IR treatment induced more severe DNA damage in both tumor cell lines than each treatment alone and also protracted the kinetics of DNA damage repair in SNB19 cells. In addition to large cell cycle disturbances, drug-IR treatment also caused depletion of the antiapoptotic proteins Akt and Raf-1 in both cell lines, along with a decrease of survivin in A549 cells in case of NVP-AUY922. The data show that simultaneous Hsp90 inhibition and irradiation may induce cell type-specific radiosensitization as well as cytotoxicity against tumor cells.
Collapse
|
29
|
Wang T, Xue Y, Wang M, Sun Q. Silencing of the hTERT gene through RNA interference induces apoptosis via bax/bcl-2 in human glioma cells. Oncol Rep 2012; 28:1153-8. [PMID: 22895663 PMCID: PMC3583528 DOI: 10.3892/or.2012.1952] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 06/21/2012] [Indexed: 12/14/2022] Open
Abstract
Glioma cells are characterized by their invasiveness and resistance to conventional therapeutics. The downregulation of human telomerase reverse transcriptase (hTERT) can lead to decreased cell proliferation and/or the induction of apoptotic cell death in cancer cells but has rarely been reported in glioma cells. Here, we assessed the effect of the silencing of the hTERT gene on cell apoptosis and its possible molecular mechanism in T98G glioma cells. We found that the silencing of the hTERT gene in T98G cells significantly decreased cell proliferation and telomerase activity, increased the number of cells in G1 phase and decreased the number of cells in S phase, and induced apoptosis via decreasing the protein level of bcl-2 and c-myc and increasing the protein levels of bax and p53.
Collapse
Affiliation(s)
- Tuo Wang
- Department of Neurosurgery, the First Affiliated Hospital, and Department of Pharmacology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, PR China
| | | | | | | |
Collapse
|
30
|
Wang YY, Sun G, Luo H, Wang XF, Lan FM, Yue X, Fu LS, Pu PY, Kang CS, Liu N, You YP. MiR-21 modulates hTERT through a STAT3-dependent manner on glioblastoma cell growth. CNS Neurosci Ther 2012; 18:722-8. [PMID: 22709411 DOI: 10.1111/j.1755-5949.2012.00349.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 04/12/2012] [Accepted: 04/15/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE As an important oncogenic miRNA, miR-21 has been reported to play crucial roles in glioblastoma (GBM) carcinogenesis. However, the precise biological function and molecular mechanism of miR-21 in GBM remain elusive. This study is designed to explore the mechanism of miR-21 involved in the control of GBM cell growth. METHODS AND RESULTS MTT assay, cell cycle analysis, and apoptosis analysis showed that reduction of miR-21 inhibited cell growth in U87 and LN229 GBM cells. Further, reduction of miR-21 decreased the expression of human telomerase reverse transcriptase (hTERT) and repressed STAT3 expression and STAT3 phosphorylation. STAT3 inhibition led to a remarkable depletion of hTERT at both mRNA and protein levels by binding to the hTERT gene promoter by performing luciferase reporter assay and chromatin Immunoprecipitation PCR. Finally, knockdown of miR-21 considerably inhibited tumor growth and diminished the expression of STAT3 and hTERT in xenograft model. CONCLUSION Our findings indicate that miR-21 regulates hTERT expression mediated by STAT3, therefore controlling GBM cell growth.
Collapse
Affiliation(s)
- Ying-Yi Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
The therapeutic function of the chemokine RANTES on the H22 hepatoma ascites model. Mol Cell Biochem 2012; 367:93-102. [PMID: 22547199 DOI: 10.1007/s11010-012-1323-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 04/18/2012] [Indexed: 12/17/2022]
Abstract
This study aimed at analyzing the therapeutic function of the chemokine RANTES on the H22 hepatoma ascites model and preliminarily explore the mechanism of RANTES in malignant ascites to provide an important reference for applying chemokines in anti-tumor therapy. The murine H22 hepatoma ascites model was used. Three treatment groups were analyzed: a RANTES treatment group, an IL-2 control group, and an NS control group. Two regimens of early treatment and late treatment were designed, and the therapeutic effect of RANTES on malignant ascites was studied by measuring changes in mouse body weight and abdominal circumference and observing the survival time. The expression of TNF-α, IFN-γ, TGF-β1, and MCP-1 in mouse ascites was detected by ELISA, and the chemotactic function of RANTES on B lymphocytes and T lymphocytes was analyzed by flow cytometry. In the early and late treatment regimens, RANTES could effectively inhibit the increase in mouse body weight and abdominal circumference in the murine H22 hepatoma ascites model. The secretion of TNF-α and IFN-γ, which had anti-tumor effects, was higher in the RANTES treatment group than in the control groups (P < 0.05), whereas the secretion of TGF-β1 and MCP-1, which promoted tumor growth, invasion, and metastasis, was lower than in the control groups (P < 0.05). RANTES had chemotactic effects on CD4(+) and CD8(+) T lymphocytes; therefore, the percentage of CD3, CD4, and CD8 in the mouse ascites in the RANTES treatment group was significantly higher than in the NS control and IL-2 treatment groups, and the CD4/CD8 ratio was also significantly higher. RANTES can effectively inhibit the increase in body weight and abdominal circumference and significantly extend survival time in mice in the H22 hepatoma ascites model.
Collapse
|
32
|
Li J, Tang B, Qu Y, Mu D. Telomerase reverse transcriptase: A novel neuroprotective mechanism involved in neonatal hypoxic‐ischemic brain injury. Int J Dev Neurosci 2011; 29:867-72. [DOI: 10.1016/j.ijdevneu.2011.07.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Revised: 07/26/2011] [Accepted: 07/26/2011] [Indexed: 01/16/2023] Open
Affiliation(s)
- Jiao Li
- Department of PediatricsWest China Second University HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Binzhi Tang
- Department of PediatricsWest China Second University HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Yi Qu
- Department of PediatricsWest China Second University HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Dezhi Mu
- Department of PediatricsWest China Second University HospitalSichuan UniversityChengduSichuan ProvinceChina
- Department of NeurologyUniversity of CaliforniaSan FranciscoCA94143USA
| |
Collapse
|
33
|
Gladych M, Wojtyla A, Rubis B. Human telomerase expression regulation. Biochem Cell Biol 2011; 89:359-76. [DOI: 10.1139/o11-037] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Since telomerase has been recognized as a relevant factor distinguishing cancer cells from normal cells, it has become a very promising target for anti-cancer therapy. A correlation between short telomere length and increased mortality was revealed in many studies. The telomerase expression/activity appears to be one of the most crucial factors to study to improve cancer therapy and prevention. However, this multisubunit enzymatic complex can be regulated at various levels. Thus, several strategies have been proposed to control telomerase in cancer cells such as anti-sense technology against TR and TERT, ribozymes against TERT, anti-estrogens, progesterone, vitamin D, retinoic acid, quadruplex stabilizers, telomere and telomerase targeting agents, modulation of interaction with other proteins involved in the regulation of telomerase and telomeres, etc. However, the transcription control of key telomerase subunits seems to play the crucial role in whole complexes activity and cancer cells immortality. Thus, the research of telomerase regulation can bring significant insight into the knowledge concerning stem cells metabolism but also ageing. This review summarizes the current state of knowledge of numerous telomerase regulation mechanisms at the transcription level in human that might become attractive anti-cancer therapy targets.
Collapse
Affiliation(s)
- Marta Gladych
- Poznan University of Medical Sciences, Department of Clinical Chemistry and Molecular Diagnostics, Przybyszewskiego 49 St., 60-355 Poznan, Poland
| | - Aneta Wojtyla
- Poznan University of Medical Sciences, Department of Clinical Chemistry and Molecular Diagnostics, Przybyszewskiego 49 St., 60-355 Poznan, Poland
| | - Blazej Rubis
- Poznan University of Medical Sciences, Department of Clinical Chemistry and Molecular Diagnostics, Przybyszewskiego 49 St., 60-355 Poznan, Poland
| |
Collapse
|
34
|
Iwami K, Natsume A, Wakabayashi T. Cytokine networks in glioma. Neurosurg Rev 2011; 34:253-63; discussion 263-4. [DOI: 10.1007/s10143-011-0320-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 12/18/2010] [Accepted: 01/28/2011] [Indexed: 12/25/2022]
|
35
|
Karmakar S, Choudhury SR, Banik NL, Ray SK. Activation of Multiple Molecular Mechanisms for Increasing Apoptosis in Human Glioblastoma T98G Xenograft. ACTA ACUST UNITED AC 2010; 2:107-113. [PMID: 21666767 DOI: 10.4172/1948-5956.1000033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Glioblastoma is the most malignant brain tumor of astroglial origin. It renders poor response or resistance to existing therapeutics. We used all-trans retinoic acid (ATRA) and interferon gamma (IFN-γ) alone and in combination for controlling human glioblastoma T98G xenografted in nude mice. Histopathological examination showed astrocytic differentiation in ATRA group, some apoptosis in IFN-γ group, and occurrence of differentiation and enhancement of apoptosis in ATRA plus IFN-γ group. ATRA plus IFN-γ induced extrinsic pathway of apoptosis by activation of caspase-8 and cleavage of Bid to tBid and also down regulation of hTERT, c-IAP2, and survivin and upregulation of Smac/Diablo to promote apoptosis. Mitochondrial release of apoptosis-inducing factor (AIF) induced caspase-independent pathway and also upregulation of calpain and caspase-dependent pathways ultimately activated caspase-3 for apoptosis. Increased activities of calpain and caspase-3 degraded 270 kD α-spectrin at the specific sites to generate 145 kD spectrin breakdown product (SBDP) and 120 kD SBDP, respectively. In situ TUNEL and double immunofluorescent labelings detected apoptosis with increased expression of calpain, caspase-12, caspase-3, and AIF in tumors after treatment with IFN-γ and most effectively with ATRA plus IFN-γ. Results indicated that ATRA plus IFN-γ activated multiple molecular mechanisms for increasing apoptosis in human glioblastoma in vivo.
Collapse
Affiliation(s)
- Surajit Karmakar
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | | | | | | |
Collapse
|