1
|
Halim CE, Deng S, Crasta KC, Yap CT. Interplay Between the Cytoskeleton and DNA Damage Response in Cancer Progression. Cancers (Basel) 2025; 17:1378. [PMID: 40282554 PMCID: PMC12025774 DOI: 10.3390/cancers17081378] [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: 03/05/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
DNA damage has emerged as a critical factor in fuelling the development and progression of cancer. DNA damage response (DDR) pathways lie at the crux of cell fate decisions following DNA damage induction, which can either trigger the repair of detrimental DNA lesions to protect cancer cells or induce the cell death machinery to eliminate damaged cells. Cytoskeletal dynamics have a critical role to play and influence the proper function of DDR pathways. Microfilaments, intermediate filaments, microtubules, and their associated proteins are well involved in the DDR. For instance, they are not only implicated in the recruitment of specific DDR molecules to the sites of DNA damage but also in the regulation of the mobility of the damaged DNA to repair sites in the periphery of the nucleus. The exquisite roles that these cytoskeletal proteins play in different DDR pathways, such as non-homologous end joining (NHEJ), homologous recombination (HR), base excision repair (BER), and nucleotide excision repair (NER), in cancer cells are extensively discussed in this review. Many cancer treatments are reliant upon inducing DNA damage in cancer cells to eliminate them; thus, it is important to shed light on factors that could affect their efficacy. Although the cytoskeleton is intricately involved in the DDR process, this has often been overlooked in cancer research and has not been exploited in developing DDR-targeting cancer therapy. Understanding the interplay between the cytoskeleton and the DDR in cancer will then provide insights into improving the development of cancer therapies that can leverage the synergistic action of DDR inhibitors and cytoskeleton-targeting agents.
Collapse
Affiliation(s)
- Clarissa Esmeralda Halim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (C.E.H.); (S.D.); (K.C.C.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (C.E.H.); (S.D.); (K.C.C.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Karen Carmelina Crasta
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (C.E.H.); (S.D.); (K.C.C.)
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Celestial T. Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (C.E.H.); (S.D.); (K.C.C.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| |
Collapse
|
2
|
Kumada T, Mimae T, Tsubokawa N, Kushitani K, Takeshima Y, Miyata Y, Okada M. Role of guanylate-binding protein 1 in the proliferation of invasive lung adenocarcinoma cells. Front Oncol 2025; 15:1434249. [PMID: 40018406 PMCID: PMC11865198 DOI: 10.3389/fonc.2025.1434249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 01/03/2025] [Indexed: 03/01/2025] Open
Abstract
Background Guanylate-binding protein 1 (GBP1) is involved in the malignant progression of lung adenocarcinoma, particularly in the acquisition of invasive potential. However, its role in tumor proliferation and therapeutic viability in invasive lung adenocarcinomas remains unclear. Methods This study included 99 patients with invasive lung adenocarcinoma, excluding those with non-invasive lepidic components, who had undergone complete pulmonary resection. Immunohistochemical staining was performed to examine the presence of GBP1, and its prognostic significance was assessed using uni- and multi-variable Cox regression analyses. Additionally, the expression levels of GBP1 gene and protein levels were evaluated in lung adenocarcinoma cell lines (PC-9, A549, NCI-H322, NCI-H441, NCI-H820, and ABC-1), and its proliferative role in these cell lines was analyzed using specific inhibitors targeting GBP1. Results GBP1 expression was detected in 45 (45.5%) patients. The 5-year overall survival rates for GBP1-positive and -negative patients were 66.0% (95% confidence interval (CI): 46.3-80.0%) and 85.7% (95% CI: 72.0-93.0%), respectively (P = 0.029). The multivariable analysis demonstrated that GBP1 positivity was an independent factor for poor overall survival (hazard ratio [HR] = 2.52 [95% CI: 1.02-6.22], P = 0.045). GBP1 gene and protein were markedly expressed in NCI-H820 than in NCI-H322 and ABC-1. The inhibitor targeting GBP1 significantly suppressed the growth of NCI-H820 but not that of NCI-H322 or ABC-1. Conclusions GBP1 is a prognostic factor that may be involved in the proliferation of invasive lung adenocarcinoma, suggesting that inhibiting GBP1 activity may be a promising therapeutic approach for lung adenocarcinoma patients expressing GBP1.
Collapse
Affiliation(s)
- Takashi Kumada
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Takahiro Mimae
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | | | - Kei Kushitani
- Department of Pathology, Hiroshima University, Hiroshima, Japan
| | - Yukio Takeshima
- Department of Pathology, Hiroshima University, Hiroshima, Japan
| | - Yoshihiro Miyata
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
3
|
Waddell S, Zhao G, Liu Z, Chen H, Zhang W, Wang Y, Miller DD, Yue J, Li W. VERU-111, an orally available tubulin inhibitor, suppresses ovarian tumor growth and metastasis. J Pharmacol Exp Ther 2025; 392:100006. [PMID: 39893008 PMCID: PMC11808928 DOI: 10.1124/jpet.124.002298] [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: 05/10/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy, with a 5-year survival rate of approximately 50%. The dismal prognosis is due in part to metastatic disease and acquired drug resistance to conventional chemotherapies such as taxanes. Colchicine binding site inhibitors (CBSIs) are attractive alternatives to taxanes because they could potentially achieve oral bioavailability and overcome drug resistance associated with the prolonged use of taxanes. VERU-111 is one of the most advanced CBSIs that is orally available, potent, and well tolerated and has shown good efficacy in several preclinical solid tumor models. Here, we demonstrate for the first time the in vitro potency of VERU-111 as well as its efficacy at inhibiting tumor growth and metastasis in an orthotopic ovarian cancer mouse model. VERU-111 has nanomolar potency against ovarian cancer cell lines and strongly inhibits colony formation, proliferation, invasion, and migration. VERU-111 disrupts microtubule formation to induce mitotic catastrophe and ultimately apoptosis in a concentration-dependent manner. The efficacy of VERU-111 was comparable with standard chemotherapy paclitaxel, the current first-line treatment of ovarian cancer, with no observed synergy with combination paclitaxel + VERU-111 treatment. In vivo, VERU-111 markedly suppressed ovarian tumor growth and completely suppressed distant organ metastasis. Together, these results support VERU-111 for its potential as a novel therapy for ovarian cancer, particularly for late-stage metastatic disease. SIGNIFICANCE STATEMENT: VERU-111 is an investigational new drug and has comparable efficacy as paclitaxel in suppressing tumor cell proliferation, colony formation, and migration in ovarian cancer models in vitro and has potent in vivo antitumor and antimetastatic activity in an orthotopic ovarian cancer mouse model. VERU-111 has low systemic toxicity and, unlike paclitaxel, is orally bioavailable and is not a substrate for the major drug efflux transporters, making it a promising and attractive alternative to taxane-based therapy.
Collapse
Affiliation(s)
- Shelby Waddell
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Guannan Zhao
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee; Department of Gynecology and Obstetrics, Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ziping Liu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee; Department of Gynecology and Obstetrics, Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Wenjing Zhang
- Genetics and Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Yaohong Wang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee; Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee.
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee; Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee.
| |
Collapse
|
4
|
Yu S, Li Y, Feng W, Zeng J, Cui X, Zhou S, Zhang P. GBP1 promotes cutaneous squamous cell carcinoma proliferation and invasion through activation of STAT3 by SP1. Exp Dermatol 2024; 33:e15112. [PMID: 38840385 DOI: 10.1111/exd.15112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/16/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
Cutaneous squamous cell carcinoma (cSCC) ranks as the second most prevalent skin tumour (excluding melanoma). However, the molecular mechanisms driving cSCC progression remain elusive. This study aimed to investigate GBP1 expression in cSCC and elucidate its potential molecular mechanisms underlying cSCC development. GBP1 expression was assessed across public databases, cell lines and tissue samples. Various assays, including clone formation, CCK8 and EdU were employed to evaluate cell proliferation, while wound healing and transwell assays determined cell migration and invasion. Subcutaneous tumour assays were conducted to assess in vivo tumour proliferation, and molecular mechanisms were explored through western blotting, immunofluorescence and immunoprecipitation. Results identified GBP1 as an oncogene in cSCC, with elevated expression in both tumour tissues and cells, strongly correlating with tumour stage and grade. In vitro and in vivo investigations revealed that increased GBP1 expression significantly enhanced cSCC cell proliferation, migration and invasion. Mechanistically, GBP1 interaction with SP1 promoted STAT3 activation, contributing to malignant behaviours. In conclusion, the study highlights the crucial role of the GBP1/SP1/STAT3 signalling axis in regulating tumour progression in cSCC. These findings provide valuable insights into the molecular mechanisms of cSCC development and offer potential therapeutic targets for interventions against cSCC.
Collapse
Affiliation(s)
- Site Yu
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yun Li
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Wenjie Feng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Jizhang Zeng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xu Cui
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Situo Zhou
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Pihong Zhang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
5
|
Tailor D, Garcia-Marques FJ, Bermudez A, Pitteri SJ, Malhotra SV. Guanylate-binding protein 1 modulates proteasomal machinery in ovarian cancer. iScience 2023; 26:108292. [PMID: 38026225 PMCID: PMC10665831 DOI: 10.1016/j.isci.2023.108292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/10/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Guanylate-binding protein 1 (GBP1) is known as an interferon-γ-induced GTPase. Here, we used genetically modified ovarian cancer (OC) cells to study the role of GBP1. The data generated show that GBP1 inhibition constrains the clonogenic potential of cancer cells. In vivo studies revealed that GBP1 overexpression in tumors promotes tumor progression and reduces median survival, whereas GBP1 inhibition delayed tumor progression with longer median survival. We employed proteomics-based thermal stability assay (CETSA) on GBP1 knockdown and overexpressed OC cells to study its molecular functions. CETSA results show that GBP1 interacts with many members of the proteasome. Furthermore, GBP1 inhibition sensitizes OC cells to paclitaxel treatment via accumulated ubiquitinylated proteins where GBP1 inhibition decreases the overall proteasomal activity. In contrast, GBP1-overexpressing cells acquired paclitaxel resistance via boosted cellular proteasomal activity. Overall, these studies expand the role of GBP1 in the activation of proteasomal machinery to acquire chemoresistance.
Collapse
Affiliation(s)
- Dhanir Tailor
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Fernando Jose Garcia-Marques
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Abel Bermudez
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sharon J. Pitteri
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sanjay V. Malhotra
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| |
Collapse
|
6
|
Ly C, Ogana H, Kim HN, Hurwitz S, Deeds EJ, Kim YM, Rowat AC. Altered physical phenotypes of leukemia cells that survive chemotherapy treatment. Integr Biol (Camb) 2023; 15:zyad006. [PMID: 37247849 DOI: 10.1093/intbio/zyad006] [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: 12/09/2022] [Revised: 04/22/2023] [Accepted: 04/29/2023] [Indexed: 05/31/2023]
Abstract
The recurrence of cancer following chemotherapy treatment is a major cause of death across solid and hematologic cancers. In B-cell acute lymphoblastic leukemia (B-ALL), relapse after initial chemotherapy treatment leads to poor patient outcomes. Here we test the hypothesis that chemotherapy-treated versus control B-ALL cells can be characterized based on cellular physical phenotypes. To quantify physical phenotypes of chemotherapy-treated leukemia cells, we use cells derived from B-ALL patients that are treated for 7 days with a standard multidrug chemotherapy regimen of vincristine, dexamethasone, and L-asparaginase (VDL). We conduct physical phenotyping of VDL-treated versus control cells by tracking the sequential deformations of single cells as they flow through a series of micron-scale constrictions in a microfluidic device; we call this method Quantitative Cyclical Deformability Cytometry. Using automated image analysis, we extract time-dependent features of deforming cells including cell size and transit time (TT) with single-cell resolution. Our findings show that VDL-treated B-ALL cells have faster TTs and transit velocity than control cells, indicating that VDL-treated cells are more deformable. We then test how effectively physical phenotypes can predict the presence of VDL-treated cells in mixed populations of VDL-treated and control cells using machine learning approaches. We find that TT measurements across a series of sequential constrictions can enhance the classification accuracy of VDL-treated cells in mixed populations using a variety of classifiers. Our findings suggest the predictive power of cell physical phenotyping as a complementary prognostic tool to detect the presence of cells that survive chemotherapy treatment. Ultimately such complementary physical phenotyping approaches could guide treatment strategies and therapeutic interventions. Insight box Cancer cells that survive chemotherapy treatment are major contributors to patient relapse, but the ability to predict recurrence remains a challenge. Here we investigate the physical properties of leukemia cells that survive treatment with chemotherapy drugs by deforming individual cells through a series of micron-scale constrictions in a microfluidic channel. Our findings reveal that leukemia cells that survive chemotherapy treatment are more deformable than control cells. We further show that machine learning algorithms applied to physical phenotyping data can predict the presence of cells that survive chemotherapy treatment in a mixed population. Such an integrated approach using physical phenotyping and machine learning could be valuable to guide patient treatments.
Collapse
Affiliation(s)
- Chau Ly
- Department of Integrative Biology & Physiology, University of California, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Heather Ogana
- Department of Pediatrics, Children's Hospital Los Angeles, Division of Hematology and Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hye Na Kim
- Department of Pediatrics, Children's Hospital Los Angeles, Division of Hematology and Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Samantha Hurwitz
- Department of Pediatrics, Children's Hospital Los Angeles, Division of Hematology and Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eric J Deeds
- Department of Integrative Biology & Physiology, University of California, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA, USA
| | - Yong-Mi Kim
- Department of Pediatrics, Children's Hospital Los Angeles, Division of Hematology and Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Amy C Rowat
- Department of Integrative Biology & Physiology, University of California, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| |
Collapse
|
7
|
Scherbakov AM, Basharina AA, Sorokin DV, Mikhaevich EI, Mizaeva IE, Mikhaylova AL, Bogush TA, Krasil’nikov MA. Targeting hormone-resistant breast cancer cells with docetaxel: a look inside the resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:103-115. [PMID: 37065867 PMCID: PMC10099602 DOI: 10.20517/cdr.2022.96] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/01/2022] [Accepted: 01/04/2023] [Indexed: 04/18/2023]
Abstract
Aim: The study aims to analyze the effect of long-term incubation of ERα-positive MCF7 breast cancer cells with 4-hydroxytamoxifen (HT) on their sensitivity to tubulin polymerization inhibitor docetaxel. Methods: The analysis of cell viability was performed by the MTT method. The expression of signaling proteins was analyzed by immunoblotting and flow cytometry. ERα activity was evaluated by gene reporter assay. To establish hormone-resistant subline MCF7, breast cancer cells were treated with 4-hydroxytamoxifen for 12 months. Results: The developed MCF7/HT subline has lost sensitivity to 4-hydroxytamoxifen, and the resistance index was 2. Increased Akt activity (2.2-fold) and decreased ERα expression (1.5-fold) were revealed in MCF7/HT cells. The activity of the estrogen receptor α was reduced (1.5-fold) in MCF7/HT. Evaluation of class III β-tubulin expression (TUBB3), a marker associated with metastasis, revealed the following trends: higher expression of TUBB3 was detected in triple-negative breast cancer MDA-MB-231 cells compared to hormone-responsive MCF7 cells (P < 0.05). The lowest expression of TUBB3 was found in hormone-resistant MCF7/HT cells (MCF7/HT < MCF7 < MDA-MB-231, approximately 1:2:4). High TUBB3 expression strongly correlated with docetaxel resistance: IC50 value of docetaxel for MDA-MB-231 cells was greater than that for MCF7 cells, whereas resistant MCF7/HT cells were the most sensitive to the drug. The accumulation of cleaved PARP (a 1.6-fold increase) and Bcl-2 downregulation (1.8-fold) were more pronounced in docetaxel-treated resistant cells (P < 0.05). The expression of cyclin D1 decreased (2.8-fold) only in resistant cells after 4 nM docetaxel treatment, while this marker was unchanged in parental MCF7 breast cancer cells. Conclusion: Further development of taxane-based chemotherapy for hormone-resistant cancer looks highly promising, especially for cancers with low TUBB3 expression.
Collapse
Affiliation(s)
- Alexander M. Scherbakov
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
- Correspondence to: Dr. Alexander M. Scherbakov, Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Kashirskoye shosse 24 bldg.15, Moscow 115522, Russia. E-mail:
| | - Anna A. Basharina
- Group of Molecular Tumor Markers, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Danila V. Sorokin
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Ekaterina I. Mikhaevich
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Iman E. Mizaeva
- Group of Molecular Tumor Markers, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Alexandra L. Mikhaylova
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Tatiana A. Bogush
- Group of Molecular Tumor Markers, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Mikhail A. Krasil’nikov
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| |
Collapse
|
8
|
Vicari HP, Lima K, Costa-Lotufo LV, Machado-Neto JA. Cellular and Molecular Effects of Eribulin in Preclinical Models of Hematologic Neoplasms. Cancers (Basel) 2022; 14:cancers14246080. [PMID: 36551566 PMCID: PMC9776580 DOI: 10.3390/cancers14246080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Despite the advances in understanding the biology of hematologic neoplasms which has resulted in the approval of new drugs, the therapeutic options are still scarce for relapsed/refractory patients. Eribulin is a unique microtubule inhibitor that is currently being used in the therapy for metastatic breast cancer and soft tissue tumors. Here, we uncover eribulin's cellular and molecular effects in a molecularly heterogeneous panel of hematologic neoplasms. Eribulin reduced cell viability and clonogenicity and promoted apoptosis and cell cycle arrest. The minimal effects of eribulin observed in the normal leukocytes suggested selectivity for malignant blood cells. In the molecular scenario, eribulin induces DNA damage and apoptosis markers. The ABCB1, ABCC1, p-AKT, p-NFκB, and NFκB levels were associated with responsiveness to eribulin in blood cancer cells, and a resistance eribulin-related target score was constructed. Combining eribulin with elacridar (a P-glycoprotein inhibitor), but not with PDTC (an NFkB inhibitor), increases eribulin-induced apoptosis in leukemia cells. In conclusion, our data indicate that eribulin leads to mitotic catastrophe and cell death in blood cancer cells. The expression and activation of MDR1, PI3K/AKT, and the NFκB-related targets may be biomarkers of the eribulin response, and the combined treatment of eribulin and elacridar may overcome drug resistance in these diseases.
Collapse
Affiliation(s)
- Hugo Passos Vicari
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulos 05508-000, Brazil
| | - Keli Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulos 05508-000, Brazil
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo 01246-903, Brazil
| | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulos 05508-000, Brazil
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulos 05508-000, Brazil
- Correspondence: ; Tel.: +55-11-3091-7467
| |
Collapse
|
9
|
Hunt EN, Kopacz JP, Vestal DJ. Unraveling the Role of Guanylate-Binding Proteins (GBPs) in Breast Cancer: A Comprehensive Literature Review and New Data on Prognosis in Breast Cancer Subtypes. Cancers (Basel) 2022; 14:cancers14112794. [PMID: 35681772 PMCID: PMC9179834 DOI: 10.3390/cancers14112794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 11/19/2022] Open
Abstract
At least one member of the Guanylate-Binding Protein (GBP) family of large interferon-induced GTPases has been classified as both a marker of good prognosis and as a potential drug target to treat breast cancers. However, the activity of individual GBPs appears to not just be tumor cell type–specific but dependent on the growth factor and/or cytokine environment in which the tumor cells reside. To clarify what we do and do not know about GBPs in breast cancer, the current literature on GBP-1, GBP-2, and GBP-5 in breast cancer has been assembled. In addition, we have analyzed the role of each of these GBPs in predicting recurrence-free survival (RFS), overall survival (OS), and distance metastasis-free survival (DMFS) as single gene products in different subtypes of breast cancers. When a large cohort of breast cancers of all types and stages were examined, GBP-1 correlated with poor RFS. However, it was the only GBP to do so. When smaller cohorts of breast cancer subtypes grouped into ER+, ER+/Her2-, and HER2+ tumors were analyzed, none of the GBPs influenced RFS, OS, or DMSF as single agents. The exception is GBP-5, which correlated with improved RFS in Her2+ breast cancers. All three GBPs individually predicted improved RFS, OS, and DMSF in ER- breast cancers, regardless of the PR or HER2 status, and TNBCs.
Collapse
|
10
|
Liu Z, Sun J, Gong T, Tang H, Shen Y, Liu C. The Prognostic and Immunological Value of Guanylate-Binding Proteins in Lower-Grade Glioma: Potential Markers or Not? Front Genet 2021; 12:651348. [PMID: 34759950 PMCID: PMC8573089 DOI: 10.3389/fgene.2021.651348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 09/23/2021] [Indexed: 11/13/2022] Open
Abstract
Seven guanylate-binding proteins (GBPs, GBP1–7), identified as a subfamily of interferon-γ-induced guanosine triphosphate hydrolases (GTPases), has been reported to be closely associated with tumor progression, metastasis, and prognosis of cancer patients in recent years. However, the expression patterns, prognostic value, immune infiltration relevance, and biological functions of GBPs in lower-grade glioma (LGG) remain elusive. In this study, by analysis and verification through multiple public data platforms, we found that GBP1, 2, 3, 4 were significantly upregulated in LGG tissues vs normal brain tissue. Analysis based on the Cox proportional hazard ratio and Kaplan–Meier plots demonstrated that the high expressions of GBP 1, 2, 3, 4 were significantly correlated with the poor prognosis of LGG patients. Correlation analysis of clinical parameters of LGG patients indicated that the expressions of GBP 1, 2, 3, 4 were significantly associated with the histological subtype and tumor histological grade of LGG. Furthermore, the correlation analysis of immune infiltration showed that the expressions of GBP1, 2, 3, 4 were significantly and positively correlated with the level of tumor immune-infiltrating cells. In particular, GBP1, 2, 3, 4 expressions were strongly correlated with the infiltration levels of monocyte, TAM, and M1/M2 macrophage, revealing their potential to regulate the polarity of macrophages. Finally, we used the GSEA method to explore the signaling pathways potentially regulated by GBP1, 2, 3, 4 and found that they were all closely associated with immune-related signaling pathways. Collectively, these findings suggested that GBP1, 2, 3, 4 were potent biomarkers to determine the prognosis and immune cell infiltration of LGG patients.
Collapse
Affiliation(s)
- Zhuang Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jifeng Sun
- Department of Radiation Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Ting Gong
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huixin Tang
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yanna Shen
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Chang Liu
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| |
Collapse
|
11
|
Szumilak M, Wiktorowska-Owczarek A, Stanczak A. Hybrid Drugs-A Strategy for Overcoming Anticancer Drug Resistance? Molecules 2021; 26:2601. [PMID: 33946916 PMCID: PMC8124695 DOI: 10.3390/molecules26092601] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Despite enormous progress in the treatment of many malignancies, the development of cancer resistance is still an important reason for cancer chemotherapy failure. Increasing knowledge of cancers' molecular complexity and mechanisms of their resistance to anticancer drugs, as well as extensive clinical experience, indicate that an effective fight against cancer requires a multidimensional approach. Multi-target chemotherapy may be achieved using drugs combination, co-delivery of medicines, or designing hybrid drugs. Hybrid drugs simultaneously targeting many points of signaling networks and various structures within a cancer cell have been extensively explored in recent years. The single hybrid agent can modulate multiple targets involved in cancer cell proliferation, possesses a simpler pharmacokinetic profile to reduce the possibility of drug interactions occurrence, and facilitates the process of drug development. Moreover, a single medication is expected to enhance patient compliance due to a less complicated treatment regimen, as well as a diminished number of adverse reactions and toxicity in comparison to a combination of drugs. As a consequence, many efforts have been made to design hybrid molecules of different chemical structures and functions as a means to circumvent drug resistance. The enormous number of studies in this field encouraged us to review the available literature and present selected research results highlighting the possible role of hybrid drugs in overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Marta Szumilak
- Department of Hospital Pharmacy, Faculty of Pharmacy, Medical University of Lodz, 1 Muszynskiego Street, 90-151 Lodz, Poland
| | - Anna Wiktorowska-Owczarek
- Department of Pharmacology and Toxicology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland;
| | - Andrzej Stanczak
- Department of Community Pharmacy, Faculty of Pharmacy, Medical University of Lodz, 1 Muszynskiego Street, 90-151 Lodz, Poland;
| |
Collapse
|
12
|
Tailor D, Resendez A, Garcia-Marques FJ, Pandrala M, Going CC, Bermudez A, Kumar V, Rafat M, Nambiar DK, Honkala A, Le QT, Sledge GW, Graves E, Pitteri SJ, Malhotra SV. Y box binding protein 1 inhibition as a targeted therapy for ovarian cancer. Cell Chem Biol 2021; 28:1206-1220.e6. [PMID: 33713600 DOI: 10.1016/j.chembiol.2021.02.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 12/29/2020] [Accepted: 02/17/2021] [Indexed: 12/15/2022]
Abstract
Y box binding protein 1 (YB-1) is a multifunctional protein associated with tumor progression and the emergence of treatment resistance (TR). Here, we report an azopodophyllotoxin small molecule, SU056, that potently inhibits tumor growth and progression via YB-1 inhibition. This YB-1 inhibitor inhibits cell proliferation, resistance to apoptosis in ovarian cancer (OC) cells, and arrests in the G1 phase. Inhibitor treatment leads to enrichment of proteins associated with apoptosis and RNA degradation pathways while downregulating spliceosome pathway. In vivo, SU056 independently restrains OC progression and exerts a synergistic effect with paclitaxel to further reduce disease progression with no observable liver toxicity. Moreover, in vitro mechanistic studies showed delayed disease progression via inhibition of drug efflux and multidrug resistance 1, and significantly lower neurotoxicity as compared with etoposide. These data suggest that YB-1 inhibition may be an effective strategy to reduce OC progression, antagonize TR, and decrease patient mortality.
Collapse
Affiliation(s)
- Dhanir Tailor
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Angel Resendez
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Fernando Jose Garcia-Marques
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Mallesh Pandrala
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Catherine C Going
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Abel Bermudez
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Vineet Kumar
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Marjan Rafat
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - Dhanya K Nambiar
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Alexander Honkala
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - George W Sledge
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Edward Graves
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sharon J Pitteri
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sanjay V Malhotra
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA; Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA.
| |
Collapse
|
13
|
Meng Y, Wang W, Chen M, Chen K, Xia X, Zhou S, Yang H. GBP1 Facilitates Indoleamine 2,3-Dioxygenase Extracellular Secretion to Promote the Malignant Progression of Lung Cancer. Front Immunol 2021; 11:622467. [PMID: 33552086 PMCID: PMC7857027 DOI: 10.3389/fimmu.2020.622467] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/04/2020] [Indexed: 01/14/2023] Open
Abstract
IDO1-mediated immune escape can lead to the malignant progression of tumors. However, the precise mechanism of IDO1 remains unclear. This study showed that IDO1 can bind to GBP1 and increase the extracellular secretion of IDO1 with the assistance of GBP1, thereby promoting the malignant proliferation and metastasis of lung cancer. In vitro study showed that the high expression levels of IDO1 and GBP1 in lung cancer cells promoted cell invasion and migration. In vivo study revealed that knock-down of IDO1 and GBP1 inhibited tumor growth and metastasis. In addition, Astragaloside IV reduces the extracellular secretion of IDO1 by blocking the interaction of IDO1 and GBP1, thereby reducing T cell exhaustion and inhibiting tumor progression. These results suggest that blocking the extracellular secretion of IDO1 may prevent T cell exhaustion and thereby enhance the effect of PD-1 inhibitors on cancer treatment.
Collapse
Affiliation(s)
- Yinnan Meng
- Laboratory of Cellular and Molecular Radiation Oncology, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, China
| | - Wei Wang
- Laboratory of Cellular and Molecular Radiation Oncology, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, China
| | - Meng Chen
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Kuifei Chen
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Xinhang Xia
- Laboratory of Cellular and Molecular Radiation Oncology, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, China
| | - Suna Zhou
- Laboratory of Cellular and Molecular Radiation Oncology, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, China
| | - Haihua Yang
- Laboratory of Cellular and Molecular Radiation Oncology, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, China.,School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
14
|
Zhang S, Shuai L, Wang D, Huang T, Yang S, Miao M, Liu F, Xu J. Pim-1 Protects Retinal Ganglion Cells by Enhancing Their Regenerative Ability Following Optic Nerve Crush. Exp Neurobiol 2020; 29:249-272. [PMID: 32624507 PMCID: PMC7344373 DOI: 10.5607/en20019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 11/19/2022] Open
Abstract
Provirus integration site Moloney murine leukemia virus (Pim-1) is a proto-oncogene reported to be associated with cell proliferation, differentiation and survival. This study was to explore the neuroprotective role of Pim-1 in a rat model subjected to optic nerve crush (ONC), and discuss its related molecules in improving the intrinsic regeneration ability of retinal ganglion cells (RGCs). Immunofluorescence staining showed that AAV2- Pim-1 infected 71% RGCs and some amacrine cells in the retina. Real-time PCR and Western blotting showed that retina infection with AAV2- Pim-1 up-regulated the Pim-1 mRNA and protein expressions compared with AAV2-GFP group. Hematoxylin-Eosin (HE) staining, γ-synuclein immunohistochemistry, Cholera toxin B (CTB) tracing and TUNEL showed that RGCs transduction with AAV2-Pim-1 prior to ONC promoted the survival of damaged RGCs and decreased cell apoptosis. RITC anterograde labeling showed that Pim-1 overexpression increased axon regeneration and promoted the recovery of visual function by pupillary light reflex and flash visual evoked potential. Western blotting showed that Pim- 1 overexpression up-regulated the expression of Stat3, p-Stat3, Akt1, p-Akt1, Akt2 and p-Akt2, as well as βIII-tubulin, GAP-43 and 4E-BP1, and downregulated the expression of SOCS1 and SOCS3, Cleaved caspase 3, Bad and Bax. These results demonstrate that Pim-1 exerted a neuroprotective effect by promoting nerve regeneration and functional recovery of RGCs. In addition, it enhanced the intrinsic regeneration capacity of RGCs after ONC by activating Stat3, Akt1 and Akt2 pathways, and inhibiting the mitochondrial apoptosis pathways. These findings suggest that Pim-1 may prove to be a potential therapeutic target for the clinical treatment of optic nerve injury.
Collapse
Affiliation(s)
- Shoumei Zhang
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Li Shuai
- Department of Health Administration, Second Military Medical University, Shanghai 200433, China
| | - Dong Wang
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| | - Tingting Huang
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| | - Shengsheng Yang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, China
| | - Mingyong Miao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, China
| | - Fang Liu
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| | - Jiajun Xu
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
15
|
MicroRNA-212-3p regulates early neurogenesis through the AKT/mTOR pathway by targeting MeCP2. Neurochem Int 2020; 137:104734. [PMID: 32246981 DOI: 10.1016/j.neuint.2020.104734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/13/2020] [Accepted: 03/28/2020] [Indexed: 11/22/2022]
Abstract
Compelling evidence has implicated role of microRNAs (miRNAs) in neurogenesis. Methyl-CpG Binding Protein 2 (MeCP2) was a key contributor to neurological disease. This study investigated whether miR-212-3p affects early neurogenesis associated with MeCP2. Microarray-based gene expression profiling of neurogenesis was employed to identify differentially expressed genes. Next, miR-212-3p expression in neural progenitor cells (NPCs) was detected using in situ hybridization and immunofluorescence. Effect of miR-212-3p and MeCP2 on cell viability, β-tubulin III expression and the AKT/mammalian target of rapamycin (mTOR) pathway activity was examined with gain- and loss-of-function experiments. In vivo experiments were also performed to verify effects of miR-212-3p on nerve tube development. MiR-212-3p expression was decreased while MeCP2 expression was increased during differentiation of NPCs. MiR-212-3p targets MeCP2 and down-regulates its expression, which resulted in repressed cell differentiation, proliferation as well as blocked AKT/mTOR pathway activation, subsequently early neurogenesis was prevented. Furthermore, overexpression of miR-212-3p inhibited nerve tube development in vivo. Taken together, miR-212-3p could restrain early neurogenesis through the blockade of AKT/mTOR pathway activation by targeting MeCP2, suggesting a promising therapeutic target for neurogenic disorders.
Collapse
|
16
|
Honkala AT, Tailor D, Malhotra SV. Guanylate-Binding Protein 1: An Emerging Target in Inflammation and Cancer. Front Immunol 2020; 10:3139. [PMID: 32117203 PMCID: PMC7025589 DOI: 10.3389/fimmu.2019.03139] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 12/24/2019] [Indexed: 12/16/2022] Open
Abstract
Guanylate-binding protein 1 (GBP1) is a large GTPase of the dynamin superfamily involved in the regulation of membrane, cytoskeleton, and cell cycle progression dynamics. In many cell types, such as endothelial cells and monocytes, GBP1 expression is strongly provoked by interferon γ (IFNγ) and acts to restrain cellular proliferation in inflammatory contexts. In immunity, GBP1 activity is crucial for the maturation of autophagosomes infected by intracellular pathogens and the cellular response to pathogen-associated molecular patterns. In chronic inflammation, GBP1 activity inhibits endothelial cell proliferation even as it protects from IFNγ-induced apoptosis. A similar inhibition of proliferation has also been found in some tumor models, such as colorectal or prostate carcinoma mouse models. However, this activity appears to be context-dependent, as in other cancers, such as oral squamous cell carcinoma and ovarian cancer, GBP1 activity appears to anchor a complex, taxane chemotherapy resistance profile where its expression levels correlate with worsened prognosis in patients. This discrepancy in GBP1 function may be resolved by GBP1's involvement in the induction of a cellular senescence phenotype, wherein anti-proliferative signals coincide with potent resistance to apoptosis and set the stage for dysregulated proliferative mechanisms present in growing cancers to hijack GBP1 as a pro- chemotherapy treatment resistance (TXR) and pro-survival factor even in the face of continued cytotoxic treatment. While the structure of GBP1 has been extensively characterized, its roles in inflammation, TXR, senescence, and other biological functions remain under-investigated, although initial findings suggest that GBP1 is a compelling target for therapeutic intervention in a variety of conditions ranging from chronic inflammatory disorders to cancer.
Collapse
Affiliation(s)
- Alexander T Honkala
- Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Dhanir Tailor
- Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Sanjay V Malhotra
- Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
17
|
Zhao J, Li X, Liu L, Cao J, Goscinski MA, Fan H, Li H, Suo Z. Oncogenic Role of Guanylate Binding Protein 1 in Human Prostate Cancer. Front Oncol 2020; 9:1494. [PMID: 31998647 PMCID: PMC6967410 DOI: 10.3389/fonc.2019.01494] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 12/11/2019] [Indexed: 01/28/2023] Open
Abstract
The Guanylate binding proteins (GBPs) are a family of large GTPases and the most studied GBP family member is the guanylate binding protein 1 (GBP1). Earlier studies revealed that GBP1 expression was inflammatory cytokines-inducible, and most of the studies focused on inflammation diseases. Increasing number of cancer studies began to reveal its biological role in cancers recently, although with contradictory findings in literature. It was discovered from our earlier prostate cancer cell line models studies that when prostate cancer cells treated with either ethidium bromide or a cell cycle inhibitor flavopiridol for a long-term, the treatment-survived tumor cells experienced metabolic reprogramming toward Warburg effect pathways with greater aggressive features, and one common finding from these cells was the upregulation of GBP1. In this study, possible role of GBP1 in two independent prostate cancer lines by application of CRISR/Cas9 gene knockout (KO) technology was investigated. The GBP1 gene KO DU145 and PC3 prostate cancer cells were significantly less aggressive in vitro, with less proliferation, migration, wound healing, and colony formation capabilities, in addition to a significantly lower level of mitochondrial oxidative phosphorylation and glycolysis. At the same time, such GBP1 KO cells were significantly more sensitive to chemotherapeutic reagents. Xenograft experiments verified a significantly slower tumor growth of the GBP1 KO cells in nude mouse model. Furthermore, GBP1 protein expression in clinical prostate cancer sample revealed its aggressive clinical feature correlation and shorter overall survival association. Collectively, our results indicate a pro-survival or oncogenic role of GBP1 in prostate cancer.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Oncology, Zhengzhou University, The Academy of Medical Science, Zhengzhou, China
| | - Xiangyu Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lan Liu
- Department of Oncology, Zhengzhou University, The Academy of Medical Science, Zhengzhou, China.,Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Cao
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mariusz Adam Goscinski
- Department of Surgery, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Huijie Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huixiang Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenhe Suo
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
18
|
Guanylate-binding protein 6 is a novel biomarker for tumorigenesis and prognosis in tongue squamous cell carcinoma. Clin Oral Investig 2019; 24:2673-2682. [PMID: 31707626 DOI: 10.1007/s00784-019-03129-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/16/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Guanylate-binding protein 6 (GBP6) is a member of the guanylate-binding protein family, and its role in cancer has not yet been reported. We aimed to investigate the clinical significance of GBP6 in oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS Next-generation sequencing was applied for analyzing differential gene expression profiling between corresponding tumor adjacent normal (CTAN) and tumor tissue from two paired OSCC patients. Real-time PCRs (RT-PCRs) were used to investigate the gene expression level of GBP6 of CTAN and tumor tissue samples from 14 TSCC patients. Immunohistochemistry was used to investigate the protein expression level of GBP6 in tumor tissues and paired CTAN tissues from 488 OSCC patients, including 183 buccal mucosa squamous cell carcinoma (BMSCC), 245 tongue squamous cell carcinoma (TSCC), and 60 lip squamous cell carcinoma (LSCC) patients. RESULTS Compared with CTAN tissues of OSCC patients, GBP6 is identified as a downregulated gene using the NGS platform, which was confirmed in 14 OSCC patients by RT-PCR. Moreover, protein expression level of GBP6 in tumor tissues was lower than that in CTAN tissues and the low GBP6 expression was correlated with poor cell differentiation/lymph node metastasis in TSCC patients. In addition, TSCC patients with low expression levels of GBP6 had poor disease-specific survival rate. CONCLUSION The low expression of GBP6 was associated with tumorigenesis and poor prognosis in OSCC patients, especially in TSCC patients. CLINICAL RELEVANCE GBP6 may serve as a novel favorable diagnostic and prognostic biomarker in TSCC patients.
Collapse
|
19
|
Non-catalytic region of tyrosine kinase adaptor protein 2 (NCK2) pathways as factor promoting aggressiveness in ovarian cancer. Int J Biol Markers 2018; 33:124-131. [PMID: 29218693 DOI: 10.5301/ijbm.5000264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND In this study we investigated the function of the non-catalytic region of tyrosine kinase adaptor protein 2 (NCK2) and its correlation with ITGB1 and ITGB4 integrins in driving ovarian cancer (OvCa) aggressiveness. We also evaluated whether NCK2 may influence prognosis in OvCa patients. METHODS Nanofluidic technology was used to analyze expression of NCK2 in 332 OvCa patients. To evaluate mRNA expression of NCK2, integrins and VEGFA in OvCa cell lines, qRT-PCR was performed. Stable NCK2 overexpression was obtained in OVCAR3. qRT-PCR and Western blot were performed to evaluate expression changes of VEGFA, vimentin, ITGB1, ITGB4, MMP2 and MMP9 under normoxia and hypoxia conditions. Coimmunoprecipitation (Co-IP) was performed in the A2780 cell line to study the interaction between NCK2 and proteins of interest. To investigate whether NCK2 can influence anchorage-independent growth, a soft agar assay was completed. Transwell invasion assay was performed on stable-transfected OVCAR-3 cell lines. RESULTS Nanofluidic data showed NCK2 can play an important role as a factor promoting tumor aggressiveness and survival in OvCa. This role was also linked to the behaviors of ITGB1 and ITGB4. Moreover, in cells overexpressing NCK2, the expression of vimentin, MMP2, MMP9, VEGFA and ITGB1, but not of ITGB4 was induced by hypoxia. Co-IP showed that NCK2 can directly bind ITGB1, but not VEGFA. NCK2 may be involved in mediating cell-extracellular matrix interactions in OvCa cells by influencing tumor aggressiveness. CONCLUSIONS This study provides evidence of a possible role of NCK2 as biomarker of OvCa progression.
Collapse
|
20
|
Kim J, Han S, Lei A, Miyano M, Bloom J, Srivastava V, Stampfer MR, Gartner ZJ, LaBarge MA, Sohn LL. Characterizing cellular mechanical phenotypes with mechano-node-pore sensing. MICROSYSTEMS & NANOENGINEERING 2018; 4:17091. [PMID: 29780657 PMCID: PMC5958920 DOI: 10.1038/micronano.2017.91] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The mechanical properties of cells change with their differentiation, chronological age, and malignant progression. Consequently, these properties may be useful label-free biomarkers of various functional or clinically relevant cell states. Here, we demonstrate mechano-node-pore sensing (mechano-NPS), a multi-parametric single-cell-analysis method that utilizes a four-terminal measurement of the current across a microfluidic channel to quantify simultaneously cell diameter, resistance to compressive deformation, transverse deformation under constant strain, and recovery time after deformation. We define a new parameter, the whole-cell deformability index (wCDI), which provides a quantitative mechanical metric of the resistance to compressive deformation that can be used to discriminate among different cell types. The wCDI and the transverse deformation under constant strain show malignant MCF-7 and A549 cell lines are mechanically distinct from non-malignant, MCF-10A and BEAS-2B cell lines, and distinguishes between cells treated or untreated with cytoskeleton-perturbing small molecules. We categorize cell recovery time, ΔTr, as instantaneous (ΔTr ~ 0 ms), transient (ΔTr ≤ 40ms), or prolonged (ΔTr > 40ms), and show that the composition of recovery types, which is a consequence of changes in cytoskeletal organization, correlates with cellular transformation. Through the wCDI and cell-recovery time, mechano-NPS discriminates between sub-lineages of normal primary human mammary epithelial cells with accuracy comparable to flow cytometry, but without antibody labeling. Mechano-NPS identifies mechanical phenotypes that distinguishes lineage, chronological age, and stage of malignant progression in human epithelial cells.
Collapse
Affiliation(s)
- Junghyun Kim
- Department of Mechanical Engineering, University of California at Berkeley, Berkeley, 94720-1740 CA USA
| | - Sewoon Han
- Department of Mechanical Engineering, University of California at Berkeley, Berkeley, 94720-1740 CA USA
| | - Andy Lei
- Department of Bioengineering, University of California at Berkeley, Berkeley, 94720-1762 CA USA
| | - Masaru Miyano
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, 91010 CA USA
| | - Jessica Bloom
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, 91010 CA USA
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, 94143 CA USA
| | - Martha R. Stampfer
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, CA, 94720 USA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, 94143 CA USA
- Graduate Program in Bioengineering, University of California, Berkeley, and
University of California, San Francisco, Berkeley, 94720 CA USA
| | - Mark A. LaBarge
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, 91010 CA USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, CA, 94720 USA
| | - Lydia L. Sohn
- Department of Mechanical Engineering, University of California at Berkeley, Berkeley, 94720-1740 CA USA
- Graduate Program in Bioengineering, University of California, Berkeley, and
University of California, San Francisco, Berkeley, 94720 CA USA
| |
Collapse
|
21
|
Quintero M, Adamoski D, Reis LMD, Ascenção CFR, Oliveira KRSD, Gonçalves KDA, Dias MM, Carazzolle MF, Dias SMG. Guanylate-binding protein-1 is a potential new therapeutic target for triple-negative breast cancer. BMC Cancer 2017; 17:727. [PMID: 29115931 PMCID: PMC5688804 DOI: 10.1186/s12885-017-3726-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 10/30/2017] [Indexed: 12/11/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is characterized by a lack of estrogen and progesterone receptor expression (ESR and PGR, respectively) and an absence of human epithelial growth factor receptor (ERBB2) amplification. Approximately 15–20% of breast malignancies are TNBC. Patients with TNBC often have an unfavorable prognosis. In addition, TNBC represents an important clinical challenge since it does not respond to hormone therapy. Methods In this work, we integrated high-throughput mRNA sequencing (RNA-Seq) data from normal and tumor tissues (obtained from The Cancer Genome Atlas, TCGA) and cell lines obtained through in-house sequencing or available from the Gene Expression Omnibus (GEO) to generate a unified list of differentially expressed (DE) genes. Methylome and proteomic data were integrated to our analysis to give further support to our findings. Genes that were overexpressed in TNBC were then curated to retain new potentially druggable targets based on in silico analysis. Knocking-down was used to assess gene importance for TNBC cell proliferation. Results Our pipeline analysis generated a list of 243 potential new targets for treating TNBC. We finally demonstrated that knock-down of Guanylate-Binding Protein 1 (GBP1 ), one of the candidate genes, selectively affected the growth of TNBC cell lines. Moreover, we showed that GBP1 expression was controlled by epidermal growth factor receptor (EGFR) in breast cancer cell lines. Conclusions We propose that GBP1 is a new potential druggable therapeutic target for treating TNBC with enhanced EGFR expression. Electronic supplementary material The online version of this article (10.1186/s12885-017-3726-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melissa Quintero
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil
| | - Douglas Adamoski
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Larissa Menezes Dos Reis
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Carolline Fernanda Rodrigues Ascenção
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Krishina Ratna Sousa de Oliveira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Kaliandra de Almeida Gonçalves
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil
| | - Marília Meira Dias
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil
| | - Marcelo Falsarella Carazzolle
- Genomic and Expression Laboratory (LGE), Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Sandra Martha Gomes Dias
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, 13083-970, Brazil.
| |
Collapse
|
22
|
Parker AL, Teo WS, McCarroll JA, Kavallaris M. An Emerging Role for Tubulin Isotypes in Modulating Cancer Biology and Chemotherapy Resistance. Int J Mol Sci 2017; 18:ijms18071434. [PMID: 28677634 PMCID: PMC5535925 DOI: 10.3390/ijms18071434] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 06/24/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022] Open
Abstract
Tubulin proteins, as components of the microtubule cytoskeleton perform critical cellular functions throughout all phases of the cell cycle. Altered tubulin isotype composition of microtubules is emerging as a feature of aggressive and treatment refractory cancers. Emerging evidence highlighting a role for tubulin isotypes in differentially influencing microtubule behaviour and broader functional networks within cells is illuminating a complex role for tubulin isotypes regulating cancer biology and chemotherapy resistance. This review focuses on the role of different tubulin isotypes in microtubule dynamics as well as in oncogenic changes that provide a survival or proliferative advantage to cancer cells within the tumour microenvironment and during metastatic processes. Consideration of the role of tubulin isotypes beyond their structural function will be essential to improving the current clinical use of tubulin-targeted chemotherapy agents and informing the development of more effective cancer therapies.
Collapse
Affiliation(s)
- Amelia L Parker
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Wee Siang Teo
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Joshua A McCarroll
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Maria Kavallaris
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
23
|
Wang W, Zhang H, Wang X, Patterson J, Winter P, Graham K, Ghosh S, Lee JC, Katsetos CD, Mackey JR, Tuszynski JA, Wong GKS, Ludueña RF. Novel mutations involving βI-, βIIA-, or βIVB-tubulin isotypes with functional resemblance to βIII-tubulin in breast cancer. PROTOPLASMA 2017; 254:1163-1173. [PMID: 27943021 DOI: 10.1007/s00709-016-1060-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/02/2016] [Indexed: 06/06/2023]
Abstract
Tubulin is the target for very widely used anti-tumor drugs, including Vinca alkaloids, taxanes, and epothilones, which are an important component of chemotherapy in breast cancer and other malignancies. Paclitaxel and other tubulin-targeting drugs bind to the β subunit of tubulin, which is a heterodimer of α and β subunits. β-Tubulin exists in the form of multiple isotypes, which are differentially expressed in normal and neoplastic cells and differ in their ability to bind to drugs. Among them, the βIII isotype is overexpressed in many aggressive and metastatic cancers and may serve as a prognostic marker in certain types of cancer. The underpinning mechanisms accounting for the overexpression of this isotype in cancer cells are unclear. To better understand the role of β-tubulin isotypes in cancer, we analyzed over 1000 clones from 90 breast cancer patients, sequencing their β-tubulin isotypes, in search of novel mutations. We have elucidated two putative emerging molecular subgroups of invasive breast cancer, each of which involve mutations in the βI-, βIIA-, or βIVB isotypes of tubulin that increase their structural, and possibly functional, resemblance to the βIII isotype. A unifying feature of the first of the two subgroups is the mutation of the highly reactive C239 residue of βI- or βIVB-tubulin to L239, R239, Y239, or P239, culminating in probable conversion of these isotypes from ROS-sensitive to ROS-resistant species. In the second subgroup, βI, βIIA, and βIVB have up to seven mutations to the corresponding residues in βIII-tubulin. Given that βIII-tubulin has emerged as a pro-survival factor, overexpression of this isotype may confer survival advantages to certain cancer cell types. In this mini-review, we bring attention to a novel mechanism by which cancer cells may undergo adaptive mutational changes involving alternate β-tubulin isotypes to make them acquire some of the pro-survival properties of βIII-tubulin. These "hybrid" tubulins, combining the sequences and/or properties of two wild-type tubulins (βIII and either βI, βIIA, or βIVB), are novel isotypes expressed solely in cancer cells and may contribute to the molecular understanding and stratification of invasive breast cancer and provide novel molecular targets for rational drug development.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Medicine, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Hangxiao Zhang
- Beijing Institute of Genomics, Key Laboratory of Genome Sciences and Information, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xumin Wang
- Beijing Institute of Genomics, Key Laboratory of Genome Sciences and Information, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jordan Patterson
- Department of Medicine, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Philip Winter
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2, Canada
| | - Kathryn Graham
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2, Canada
| | - Sunita Ghosh
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2, Canada
| | - John C Lee
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Christos D Katsetos
- Department of Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, 19134, USA
- Department of Pathology and Laboratory Medicine, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, 19134, USA
| | - John R Mackey
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2, Canada
| | - Jack A Tuszynski
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2, Canada
| | - Gane Ka-Shu Wong
- Department of Medicine, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Richard F Ludueña
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
24
|
Lan Q, Wang A, Cheng Y, Mukasa A, Ma J, Hong L, Yu S, Sun L, Huang Q, Purow B, Li M. Guanylate binding protein-1 mediates EGFRvIII and promotes glioblastoma growth in vivo but not in vitro. Oncotarget 2016; 7:9680-91. [PMID: 26848767 PMCID: PMC4891076 DOI: 10.18632/oncotarget.7109] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 01/22/2016] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and deadly primary brain tumor in adults. Epidermal growth factor receptor (EGFR) is frequently amplified and mutated in GBM. We previously reported that Guanylate binding protein-1 (GBP1) is a novel transcriptional target gene of EGFR and plays a role in GBM invasion. Here we demonstrate that GBP1 can also be induced by EGFRvIII at the transcriptional level through the p38 MAPK/Yin Yang 1 (YY1) signaling pathway. Silencing of GBP1 by RNA interference significantly inhibits EGFRvIII-mediated GBM cell proliferation in vitro and in a mouse model. Overexpression of GBP1 has no obvious effect on glioblastoma cell proliferation in vitro. In contrast, in an orthotopic glioma mouse model GBP1 overexpression significantly promotes glioma growth and reduces survival rate of glioma-bearing mice by increasing cell proliferation and decreasing cell apoptosis in tumor. Clinically, GBP1 expression is elevated in human GBM tumors and positively correlates with EGFRvIII status in GBM specimens, and its expression is inversely correlated with the survival rate of GBM patients. Taken together, these results reveal that GBP1 may serve as a potential therapeutic target for GBMs with EGFRvIII mutation.
Collapse
Affiliation(s)
- Qing Lan
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Aidong Wang
- The Experimental Center, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yanwei Cheng
- Department of Life Sciences, Luoyang Normal University, Luoyang, Henan Province, China
| | - Akitaki Mukasa
- Department of Neurosurgery, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Jiawei Ma
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Lei Hong
- The Experimental Center, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Shuye Yu
- The Experimental Center, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Lili Sun
- The Experimental Center, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qiang Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Benjamin Purow
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Ming Li
- The Experimental Center, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,Department of Neurology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
25
|
Wadi S, Tipton AR, Trendel JA, Khuder SA, Vestal DJ. hGBP-1 Expression Predicts Shorter Progression-Free Survival in Ovarian Cancers, While Contributing to Paclitaxel Resistance. ACTA ACUST UNITED AC 2016; 7:994-1007. [PMID: 28090373 PMCID: PMC5226657 DOI: 10.4236/jct.2016.713097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Ovarian cancer is the gynecological cancer with the poorest prognosis. One significant reason is the development of resistance to the chemotherapeutic drugs used in its treatment. The large GTPase, hGBP-1, has been implicated in paclitaxel resistance in ovarian cell lines. Forced expression of hGBP-1 in SKOV3 ovarian cancer cells protects them from paclitaxel-induced cell death. However, prior to this study, nothing was known about whether hGBP-1 was expressed in ovarian tumors and whether its expression correlated with paclitaxel resistance. hGBP-1 is expressed in 17% of ovarian tumors from patients that have not yet received treatment. However, at least 80% of the ovarian tumors that recurred after therapies that included a tax-ane, either paclitaxel or docetaxel, were positive for hGBP-1. In addition, hGBP-1 expression predicts a significantly shorter progression-free survival in ovarian cancers. Based on these studies, hGBP-1 could prove to be a potential biomarker for paclitaxel resistance in ovarian cancer.
Collapse
Affiliation(s)
- Suzan Wadi
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Aaron R Tipton
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Jill A Trendel
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Sadik A Khuder
- Department of Medicine, University of Toledo, Toledo, OH, USA
| | - Deborah J Vestal
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|
26
|
|
27
|
Guanylate-Binding Protein-1 protects ovarian cancer cell lines but not breast cancer cell lines from killing by paclitaxel. Biochem Biophys Res Commun 2016; 478:1617-23. [PMID: 27590579 DOI: 10.1016/j.bbrc.2016.08.169] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 08/30/2016] [Indexed: 12/14/2022]
Abstract
Forced expression of the cytokine-induced large GTPase, human Guanylate-Binding Protein-1 (hGBP-1), in ovarian cancer cell lines increases resistance to paclitaxel. Elevated hGBP-1 RNA in ovarian tumors correlates with shorter recurrence-free survival. In contract, hGBP-1 is part of a gene signature predicting improved prognosis in all subtypes of breast cancers. hGBP-1 does not confer paclitaxel resistance on MCF-7 and TMX2-28 breast cancer cells. Expression of the isotype of the hGBP-1-interacting protein, PIM1, which may contribute to paclitaxel resistance when associated with hGBP-1, is different in breast and ovarian cancer cell lines. Breast cancer cell lines express the 44 kDa isoform of PIM-1, and ovarian cancer cell lines express the 33 kDa isoform.
Collapse
|
28
|
Britzen-Laurent N, Herrmann C, Naschberger E, Croner RS, Stürzl M. Pathophysiological role of guanylate-binding proteins in gastrointestinal diseases. World J Gastroenterol 2016; 22:6434-6443. [PMID: 27605879 PMCID: PMC4968125 DOI: 10.3748/wjg.v22.i28.6434] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/25/2016] [Accepted: 06/13/2016] [Indexed: 02/06/2023] Open
Abstract
Guanylate-binding proteins (GBPs) are interferon-stimulated factors involved in the defense against cellular pathogens and inflammation. These proteins, particularly GBP-1, the most prominent member of the family, have been established as reliable markers of interferon-γ-activated cells in various diseases, including colorectal carcinoma (CRC) and inflammatory bowel diseases (IBDs). In CRC, GBP-1 expression is associated with a Th1-dominated angiostatic micromilieu and is correlated with a better outcome. Inhibition of tumor growth by GBP-1 is the result of its strong anti-angiogenic activity as well as its direct anti-tumorigenic effect on tumor cells. In IBD, GBP-1 mediates the anti-proliferative effects of interferon-γ on intestinal epithelial cells. In addition, it plays a protective role on the mucosa by preventing cell apoptosis, by inhibiting angiogenesis and by regulating the T-cell receptor signaling. These functions rely to a large extent on the ability of GBP-1 to interact with and remodel the actin cytoskeleton.
Collapse
|
29
|
Persico M, Di Dato A, Orteca N, Cimino P, Novellino E, Fattorusso C. Use of Integrated Computational Approaches in the Search for New Therapeutic Agents. Mol Inform 2016; 35:309-25. [DOI: 10.1002/minf.201501028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 06/21/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Marco Persico
- Department of Pharmacy, University of Naples “Federico II”; Via D. Montesano 49 80131 Napoli Italy
- Italian Malaria Network - Centro Interuniversitario di Ricerche Sulla Malaria (CIRM); Department of Experimental Medicine and Biochemical Sciences; Via Del Giochetto 06126 Perugia Italy
| | - Antonio Di Dato
- Department of Pharmacy, University of Naples “Federico II”; Via D. Montesano 49 80131 Napoli Italy
- Italian Malaria Network - Centro Interuniversitario di Ricerche Sulla Malaria (CIRM); Department of Experimental Medicine and Biochemical Sciences; Via Del Giochetto 06126 Perugia Italy
| | - Nausicaa Orteca
- Department of Pharmacy, University of Naples “Federico II”; Via D. Montesano 49 80131 Napoli Italy
- Italian Malaria Network - Centro Interuniversitario di Ricerche Sulla Malaria (CIRM); Department of Experimental Medicine and Biochemical Sciences; Via Del Giochetto 06126 Perugia Italy
| | - Paola Cimino
- Department of Pharmacy; University of Salerno; Via Giovanni Paolo II 132 84084 Fisciano, Salerno Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples “Federico II”; Via D. Montesano 49 80131 Napoli Italy
| | - Caterina Fattorusso
- Department of Pharmacy, University of Naples “Federico II”; Via D. Montesano 49 80131 Napoli Italy
- Italian Malaria Network - Centro Interuniversitario di Ricerche Sulla Malaria (CIRM); Department of Experimental Medicine and Biochemical Sciences; Via Del Giochetto 06126 Perugia Italy
| |
Collapse
|
30
|
van Neerven SM, Tieken M, Vermeulen L, Bijlsma MF. Bidirectional interconversion of stem and non-stem cancer cell populations: A reassessment of theoretical models for tumor heterogeneity. Mol Cell Oncol 2015; 3:e1098791. [PMID: 27308617 PMCID: PMC4905404 DOI: 10.1080/23723556.2015.1098791] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/18/2015] [Accepted: 09/18/2015] [Indexed: 02/07/2023]
Abstract
Resolving the origin of intratumor heterogeneity has proven to be one of the central challenges in cancer research during recent years. Two theoretical models explaining the emergence of intratumor heterogeneity have come to dominate cancer biology literature: the clonal evolution model and the hierarchical/cancer stem cell model. Recently, a plastic model that combines elements of both the clonal and the hierarchical model has gained traction. Basically, this model proposes that cancer stem cells engage in bidirectional interconversion with non-stem cells, thereby providing the missing link between the 2 conventional models. Confirming bidirectional interconversion as a hallmark of cancer is a crucial step in understanding tumor heterogeneity and has important therapeutic implications. In this review, current methodologies and theoretical and empirical evidence regarding bidirectional interconversion will be discussed.
Collapse
Affiliation(s)
- Sanne M van Neerven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academic Medical Center , Amsterdam, The Netherlands
| | - Mathijs Tieken
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academic Medical Center , Amsterdam, The Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academic Medical Center , Amsterdam, The Netherlands
| | - Maarten F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academic Medical Center , Amsterdam, The Netherlands
| |
Collapse
|
31
|
Stabilizing versus destabilizing the microtubules: a double-edge sword for an effective cancer treatment option? Anal Cell Pathol (Amst) 2015; 2015:690916. [PMID: 26484003 PMCID: PMC4592889 DOI: 10.1155/2015/690916] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/29/2015] [Accepted: 09/01/2015] [Indexed: 11/17/2022] Open
Abstract
Microtubules are dynamic and structural cellular components involved in several cell functions, including cell shape, motility, and intracellular trafficking. In proliferating cells, they are essential components in the division process through the formation of the mitotic spindle. As a result of these functions, tubulin and microtubules are targets for anticancer agents. Microtubule-targeting agents can be divided into two groups: microtubule-stabilizing, and microtubule-destabilizing agents. The former bind to the tubulin polymer and stabilize microtubules, while the latter bind to the tubulin dimers and destabilize microtubules. Alteration of tubulin-microtubule equilibrium determines the disruption of the mitotic spindle, halting the cell cycle at the metaphase-anaphase transition and, eventually, resulting in cell death. Clinical application of earlier microtubule inhibitors, however, unfortunately showed several limits, such as neurological and bone marrow toxicity and the emergence of drug-resistant tumor cells. Here we review several natural and synthetic microtubule-targeting agents, which showed antitumor activity and increased efficacy in comparison to traditional drugs in various preclinical and clinical studies. Cryptophycins, combretastatins, ombrabulin, soblidotin, D-24851, epothilones and discodermolide were used in clinical trials. Some of them showed antiangiogenic and antivascular activity and others showed the ability to overcome multidrug resistance, supporting their possible use in chemotherapy.
Collapse
|
32
|
Mariani M, Karki R, Spennato M, Pandya D, He S, Andreoli M, Fiedler P, Ferlini C. Class III β-tubulin in normal and cancer tissues. Gene 2015; 563:109-14. [PMID: 25839941 DOI: 10.1016/j.gene.2015.03.061] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 12/13/2022]
Abstract
Microtubules are polymeric structures composed of tubulin subunits. Each subunit consists of a heterodimer of α- and β-tubulin. At least seven β-tubulin isotypes, or classes, have been identified in human cells, and constitutive isotype expression appears to be tissue specific. Class III β-tubulin (βIII-tubulin) expression is normally confined to testes and tissues derived from neural cristae. However, its expression can be induced in other tissues, both normal and neoplastic, subjected to a toxic microenvironment characterized by hypoxia and poor nutrient supply. In this review, we will summarize the mechanisms underlying βIII-tubulin constitutive and induced expression. We will also illustrate its capacity to serve as a biomarker of neural commitment in normal tissues and as a pure prognostic biomarker in cancer patients.
Collapse
Affiliation(s)
| | - Roshan Karki
- Danbury Hospital Research Institute, Danbury, CT, USA
| | | | - Deep Pandya
- Danbury Hospital Research Institute, Danbury, CT, USA
| | - Shiquan He
- Danbury Hospital Research Institute, Danbury, CT, USA
| | | | - Paul Fiedler
- Danbury Hospital Research Institute, Danbury, CT, USA
| | | |
Collapse
|
33
|
Das V, Štěpánková J, Hajdúch M, Miller JH. Role of tumor hypoxia in acquisition of resistance to microtubule-stabilizing drugs. Biochim Biophys Acta Rev Cancer 2015; 1855:172-82. [DOI: 10.1016/j.bbcan.2015.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/12/2015] [Accepted: 02/01/2015] [Indexed: 12/19/2022]
|
34
|
Katsetos CD, Reginato MJ, Baas PW, D'Agostino L, Legido A, Tuszyn Ski JA, Dráberová E, Dráber P. Emerging microtubule targets in glioma therapy. Semin Pediatr Neurol 2015; 22:49-72. [PMID: 25976261 DOI: 10.1016/j.spen.2015.03.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Major advances in the genomics and epigenomics of diffuse gliomas and glioblastoma to date have not been translated into effective therapy, necessitating pursuit of alternative treatment approaches for these therapeutically challenging tumors. Current knowledge of microtubules in cancer and the development of new microtubule-based treatment strategies for high-grade gliomas are the topic in this review article. Discussed are cellular, molecular, and pharmacologic aspects of the microtubule cytoskeleton underlying mitosis and interactions with other cellular partners involved in cell cycle progression, directional cell migration, and tumor invasion. Special focus is placed on (1) the aberrant overexpression of βIII-tubulin, a survival factor associated with hypoxic tumor microenvironment and dynamic instability of microtubules; (2) the ectopic overexpression of γ-tubulin, which in addition to its conventional role as a microtubule-nucleating protein has recently emerged as a transcription factor interacting with oncogenes and kinases; (3) the microtubule-severing ATPase spastin and its emerging role in cell motility of glioblastoma cells; and (4) the modulating role of posttranslational modifications of tubulin in the context of interaction of microtubules with motor proteins. Specific antineoplastic strategies discussed include downregulation of targeted molecules aimed at achieving a sensitization effect on currently used mainstay therapies. The potential role of new classes of tubulin-binding agents and ATPase inhibitors is also examined. Understanding the cellular and molecular mechanisms underpinning the distinct behaviors of microtubules in glioma tumorigenesis and drug resistance is key to the discovery of novel molecular targets that will fundamentally change the prognostic outlook of patients with diffuse high-grade gliomas.
Collapse
Affiliation(s)
- Christos D Katsetos
- Department of Pediatrics, Drexel University College of Medicine, Section of Neurology and Pediatric Neuro-oncology Program, St Christopher's Hospital for Children, Philadelphia, PA; Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, PA.
| | - Mauricio J Reginato
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA
| | - Peter W Baas
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA
| | - Luca D'Agostino
- Department of Pediatrics, Drexel University College of Medicine, Section of Neurology and Pediatric Neuro-oncology Program, St Christopher's Hospital for Children, Philadelphia, PA
| | - Agustin Legido
- Department of Pediatrics, Drexel University College of Medicine, Section of Neurology and Pediatric Neuro-oncology Program, St Christopher's Hospital for Children, Philadelphia, PA
| | - Jack A Tuszyn Ski
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada; Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Eduarda Dráberová
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Pavel Dráber
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
35
|
McCarroll JA, Sharbeen G, Liu J, Youkhana J, Goldstein D, McCarthy N, Limbri LF, Dischl D, Ceyhan GO, Erkan M, Johns AL, Biankin AV, Kavallaris M, Phillips PA. βIII-tubulin: a novel mediator of chemoresistance and metastases in pancreatic cancer. Oncotarget 2015; 6:2235-49. [PMID: 25544769 PMCID: PMC4385848 DOI: 10.18632/oncotarget.2946] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/09/2014] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer is a leading cause of cancer-related deaths in Western societies. This poor prognosis is due to chemotherapeutic drug resistance and metastatic spread. Evidence suggests that microtubule proteins namely, β-tubulins are dysregulated in tumor cells and are involved in regulating chemosensitivity. However, the role of β-tubulins in pancreatic cancer are unknown. We measured the expression of different β-tubulin isotypes in pancreatic adenocarcinoma tissue and pancreatic cancer cells. Next, we used RNAi to silence βIII-tubulin expression in pancreatic cancer cells, and measured cell growth in the absence and presence of chemotherapeutic drugs. Finally, we assessed the role of βIII-tubulin in regulating tumor growth and metastases using an orthotopic pancreatic cancer mouse model. We found that βIII-tubulin is highly expressed in pancreatic adenocarcinoma tissue and pancreatic cancer cells. Further, we demonstrated that silencing βIII-tubulin expression reduced pancreatic cancer cell growth and tumorigenic potential in the absence and presence of chemotherapeutic drugs. Finally, we demonstrated that suppression of βIII-tubulin reduced tumor growth and metastases in vivo. Our novel data demonstrate that βIII-tubulin is a key player in promoting pancreatic cancer growth and survival, and silencing its expression may be a potential therapeutic strategy to increase the long-term survival of pancreatic cancer patients.
Collapse
Affiliation(s)
- Joshua A. McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Australia, Sydney, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW, Australia
| | - George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales (UNSW Australia), Sydney, Australia
| | - Jie Liu
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales (UNSW Australia), Sydney, Australia
| | - Janet Youkhana
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales (UNSW Australia), Sydney, Australia
| | - David Goldstein
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales (UNSW Australia), Sydney, Australia
- Prince of Wales Hospital, Prince of Wales Clinical School, Sydney, NSW, Australia
| | - Nigel McCarthy
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Australia, Sydney, Australia
| | - Lydia F. Limbri
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales (UNSW Australia), Sydney, Australia
| | - Dominic Dischl
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Güralp O. Ceyhan
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Mert Erkan
- Department of Surgery Koc University School of Medicine, Istanbul, Turkey
| | - Amber L. Johns
- The Kinghorn Cancer Centre, Cancer Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - Andrew V. Biankin
- The Kinghorn Cancer Centre, Cancer Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow, Scotland G61 1BD, United Kingdom
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Australia, Sydney, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW, Australia
| | - Phoebe A. Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales (UNSW Australia), Sydney, Australia
| |
Collapse
|
36
|
Pandya D, Mariani M, McHugh M, Andreoli M, Sieber S, He S, Dowell-Martino C, Fiedler P, Scambia G, Ferlini C. Herpes virus microRNA expression and significance in serous ovarian cancer. PLoS One 2014; 9:e114750. [PMID: 25485872 PMCID: PMC4259392 DOI: 10.1371/journal.pone.0114750] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 11/13/2014] [Indexed: 12/23/2022] Open
Abstract
Serous ovarian cancer (SEOC) is the deadliest gynecologic malignancy. MicroRNAs (miRNAs) are a class of small noncoding RNAs which regulate gene expression and protein translation. MiRNAs are also encoded by viruses with the intent of regulating their own genes and those of the infected cells. This is the first study assessing viral miRNAs in SEOC. MiRNAs sequencing data from 487 SEOC patients were downloaded from the TCGA website and analyzed through in-house sequencing pipeline. To cross-validate TCGA analysis, we measured the expression of miR-H25 by quantitative immunofluorescence in an additional cohort of 161 SEOC patients. Gene, miRNA expression, and cytotoxicity assay were performed on multiple ovarian cancer cell lines transfected with miR-H25 and miR-BART7. Outcome analysis was performed using multivariate Cox and Kaplan-Meier method. Viral miRNAs are more expressed in SEOC than in normal tissues. Moreover, Herpetic viral miRNAs (miR-BART7 from EBV and miR-H25 from HSV-2) are significant and predictive biomarkers of outcome in multivariate Cox analysis. MiR-BART7 correlates with resistance to first line chemotherapy and early death, whereas miR-H25 appears to impart a protective effect and long term survival. Integrated analysis of gene and viral miRNAs expression suggests that miR-BART7 induces directly cisplatin-resistance, while miR-H25 alters RNA processing and affects the expression of noxious human miRNAs such as miR-143. This is the first investigation linking viral miRNA expression to ovarian cancer outcome. Viral miRNAs can be useful to develop biomarkers for early diagnosis and as a potential therapeutic tool to reduce SEOC lethality.
Collapse
Affiliation(s)
- Deep Pandya
- Danbury Hospital Research Institute, Danbury, CT, United States of America
| | - Marisa Mariani
- Danbury Hospital Research Institute, Danbury, CT, United States of America
| | - Mark McHugh
- Danbury Hospital Research Institute, Danbury, CT, United States of America
| | - Mirko Andreoli
- Danbury Hospital Research Institute, Danbury, CT, United States of America
| | - Steven Sieber
- Danbury Hospital Research Institute, Danbury, CT, United States of America
| | - Shiquan He
- Danbury Hospital Research Institute, Danbury, CT, United States of America
| | | | - Paul Fiedler
- Danbury Hospital Research Institute, Danbury, CT, United States of America
| | - Giovanni Scambia
- Department of Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Cristiano Ferlini
- Danbury Hospital Research Institute, Danbury, CT, United States of America
- * E-mail:
| |
Collapse
|
37
|
McCarroll JA, Gan PP, Erlich RB, Liu M, Dwarte T, Sagnella SS, Akerfeldt MC, Yang L, Parker AL, Chang MH, Shum MS, Byrne FL, Kavallaris M. TUBB3/βIII-tubulin acts through the PTEN/AKT signaling axis to promote tumorigenesis and anoikis resistance in non-small cell lung cancer. Cancer Res 2014; 75:415-25. [PMID: 25414139 DOI: 10.1158/0008-5472.can-14-2740] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
βIII-tubulin (encoded by TUBB3) expression is associated with therapeutic resistance and aggressive disease in non-small cell lung cancer (NSCLC), but the basis for its pathogenic influence is not understood. Functional and differential proteomics revealed that βIII-tubulin regulates expression of proteins associated with malignant growth and metastases. In particular, the adhesion-associated tumor suppressor maspin was differentially regulated by βIII-tubulin. Functionally, βIII-tubulin suppression altered cell morphology, reduced tumor spheroid outgrowth, and increased sensitivity to anoikis. Mechanistically, the PTEN/AKT signaling axis was defined as a critical pathway regulated by βIII-tubulin in NSCLC cells. βIII-Tubulin blockage in vivo reduced tumor incidence and growth. Overall, our findings revealed how βIII-tubulin influences tumor growth in NSCLC, defining new biologic functions and mechanism of action of βIII-tubulin in tumorigenesis.
Collapse
Affiliation(s)
- Joshua A McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia. ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales Australia, New South Wales, Australia
| | - Pei Pei Gan
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Rafael B Erlich
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Marjorie Liu
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Tanya Dwarte
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Sharon S Sagnella
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia. ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales Australia, New South Wales, Australia
| | - Mia C Akerfeldt
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Lu Yang
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Amelia L Parker
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Melissa H Chang
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Michael S Shum
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Frances L Byrne
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, University of New South Wales Australia, Sydney, New South Wales, Australia. ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales Australia, New South Wales, Australia.
| |
Collapse
|
38
|
Bordji K, Grandval A, Cuhna-Alves L, Lechapt-Zalcman E, Bernaudin M. Hypoxia-inducible factor-2α (HIF-2α), but not HIF-1α, is essential for hypoxic induction of class III β-tubulin expression in human glioblastoma cells. FEBS J 2014; 281:5220-36. [DOI: 10.1111/febs.13062] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 08/29/2014] [Accepted: 09/18/2014] [Indexed: 01/05/2023]
Affiliation(s)
- Karim Bordji
- CNRS; UMR 6301 ISTCT; CERVOxy group; GIP Cyceron; Caen France
- CEA; DSV/I2BM; UMR 6301 ISTCT; Caen France
- Université de Basse-Normandie; UMR 6301 ISTCT; Caen France
- Normandie University; Caen France
| | - Alexandra Grandval
- CNRS; UMR 6301 ISTCT; CERVOxy group; GIP Cyceron; Caen France
- CEA; DSV/I2BM; UMR 6301 ISTCT; Caen France
- Université de Basse-Normandie; UMR 6301 ISTCT; Caen France
- Normandie University; Caen France
| | - Leilane Cuhna-Alves
- CNRS; UMR 6301 ISTCT; CERVOxy group; GIP Cyceron; Caen France
- CEA; DSV/I2BM; UMR 6301 ISTCT; Caen France
- Université de Basse-Normandie; UMR 6301 ISTCT; Caen France
- Normandie University; Caen France
| | - Emmanuèle Lechapt-Zalcman
- CNRS; UMR 6301 ISTCT; CERVOxy group; GIP Cyceron; Caen France
- CEA; DSV/I2BM; UMR 6301 ISTCT; Caen France
- Université de Basse-Normandie; UMR 6301 ISTCT; Caen France
- Normandie University; Caen France
- CHU de Caen; Service d'Anatomie et Cytologie Pathologique; Caen France
| | - Myriam Bernaudin
- CNRS; UMR 6301 ISTCT; CERVOxy group; GIP Cyceron; Caen France
- CEA; DSV/I2BM; UMR 6301 ISTCT; Caen France
- Université de Basse-Normandie; UMR 6301 ISTCT; Caen France
- Normandie University; Caen France
| |
Collapse
|
39
|
Andreoli M, Persico M, Kumar A, Orteca N, Kumar V, Pepe A, Mahalingam S, Alegria A, Petrella L, Sevciunaite L, Camperchioli A, Mariani M, Di Dato A, Novellino E, Scambia G, Malhotra SV, Ferlini C, Fattorusso C. Identification of the first inhibitor of the GBP1:PIM1 interaction. Implications for the development of a new class of anticancer agents against paclitaxel resistant cancer cells. J Med Chem 2014; 57:7916-32. [PMID: 25211704 PMCID: PMC4191604 DOI: 10.1021/jm5009902] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Indexed: 01/16/2023]
Abstract
Class III β-tubulin plays a prominent role in the development of drug resistance to paclitaxel by allowing the incorporation of the GBP1 GTPase into microtubules. Once in the cytoskeleton, GBP1 binds to prosurvival kinases such as PIM1 and initiates a signaling pathway that induces resistance to paclitaxel. Therefore, the inhibition of the GBP1:PIM1 interaction could potentially revert resistance to paclitaxel. A panel of 44 4-azapodophyllotoxin derivatives was screened in the NCI-60 cell panel. The result is that 31 are active and the comparative analysis demonstrated specific activity in paclitaxel-resistant cells. Using surface plasmon resonance, we were able to prove that NSC756093 is a potent in vitro inhibitor of the GBP1:PIM1 interaction and that this property is maintained in vivo in ovarian cancer cells resistant to paclitaxel. Through bioinformatics, molecular modeling, and mutagenesis studies, we identified the putative NSC756093 binding site at the interface between the helical and the LG domain of GBP1. According to our results by binding to this site, the NSC756093 compound is able to stabilize a conformation of GBP1 not suitable for binding to PIM1.
Collapse
Affiliation(s)
- Mirko Andreoli
- Danbury Hospital Research Institute, 24 Hospital Avenue, Danbury, Connecticut 06810, United States
| | - Marco Persico
- Department
of Pharmacy, University of Napoli “Federico
II”, Via D. Montesano
49, 80131 Napoli, Italy
| | - Ajay Kumar
- School of Environmental Affairs, Universidad Metropolitana, Avenue Ana G. Mèndez, San Juan, Puerto Rico PR 00928, United States
| | - Nausicaa Orteca
- Department
of Pharmacy, University of Napoli “Federico
II”, Via D. Montesano
49, 80131 Napoli, Italy
| | - Vineet Kumar
- Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National
Laboratory for Cancer Research, 1050 Boyles Street, Frederick, Maryland 21702, United States
| | - Antonella Pepe
- Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National
Laboratory for Cancer Research, 1050 Boyles Street, Frederick, Maryland 21702, United States
| | - Sakkarapalayam Mahalingam
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United
States
| | - Antonio
E. Alegria
- Department
of Chemistry, University of Puerto Rico
at Humacao, 100 Road
908, Humacao, Puerto Rico PR 00791, United States
| | - Lella Petrella
- Laboratory
of Molecular Oncology, Jean Paul II Research Foundation, Largo A. Gemelli 1, 86100 Campobasso, Italy
| | - Laima Sevciunaite
- Laboratory
of Molecular Oncology, Jean Paul II Research Foundation, Largo A. Gemelli 1, 86100 Campobasso, Italy
| | - Alessia Camperchioli
- Laboratory
of Molecular Oncology, Jean Paul II Research Foundation, Largo A. Gemelli 1, 86100 Campobasso, Italy
| | - Marisa Mariani
- Danbury Hospital Research Institute, 24 Hospital Avenue, Danbury, Connecticut 06810, United States
| | - Antonio Di Dato
- Department
of Pharmacy, University of Napoli “Federico
II”, Via D. Montesano
49, 80131 Napoli, Italy
| | - Ettore Novellino
- Department
of Pharmacy, University of Napoli “Federico
II”, Via D. Montesano
49, 80131 Napoli, Italy
| | - Giovanni Scambia
- Department of Obstetrics
and Gynecology, Catholic University of the
Sacred Heart, Largo A.
Gemelli 8, 00168 Roma, Italy
| | - Sanjay V. Malhotra
- Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National
Laboratory for Cancer Research, 1050 Boyles Street, Frederick, Maryland 21702, United States
| | - Cristiano Ferlini
- Danbury Hospital Research Institute, 24 Hospital Avenue, Danbury, Connecticut 06810, United States
| | - Caterina Fattorusso
- Department
of Pharmacy, University of Napoli “Federico
II”, Via D. Montesano
49, 80131 Napoli, Italy
| |
Collapse
|
40
|
Fukumoto M, Amanuma T, Kuwahara Y, Shimura T, Suzuki M, Mori S, Kumamoto H, Saito Y, Ohkubo Y, Duan Z, Sano K, Oguchi T, Kainuma K, Usami S, Kinoshita K, Lee I, Fukumoto M. Guanine nucleotide-binding protein 1 is one of the key molecules contributing to cancer cell radioresistance. Cancer Sci 2014; 105:1351-9. [PMID: 25098609 PMCID: PMC4462352 DOI: 10.1111/cas.12489] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 07/18/2014] [Accepted: 07/22/2014] [Indexed: 01/07/2023] Open
Abstract
Standard fractionated radiotherapy for the treatment of cancer consists of daily irradiation of 2-Gy X-rays, 5 days a week for 5-8 weeks. To understand the characteristics of radioresistant cancer cells and to develop more effective radiotherapy, we established a series of novel, clinically relevant radioresistant (CRR) cells that continue to proliferate with 2-Gy X-ray exposure every 24 h for more than 30 days in vitro. We studied three human and one murine cell line, and their CRR derivatives. Guanine nucleotide-binding protein 1 (GBP1) gene expression was higher in all CRR cells than their corresponding parental cells. GBP1 knockdown by siRNA cancelled radioresistance of CRR cells in vitro and in xenotransplanted tumor tissues in nude mice. The clinical relevance of GBP1 was immunohistochemically assessed in 45 cases of head and neck cancer tissues. Patients with GBP1-positive cancer tended to show poorer response to radiotherapy. We recently reported that low dose long-term fractionated radiation concentrates cancer stem cells (CSCs). Immunofluorescence staining of GBP1 was stronger in CRR cells than in corresponding parental cells. The frequency of Oct4-positive CSCs was higher in CRR cells than in parental cells, however, was not as common as GBP1-positive cells. GBP1-positive cells were radioresistant, but radioresistant cells were not necessarily CSCs. We concluded that GBP1 overexpression is necessary for the radioresistant phenotype in CRR cells, and that targeting GBP1-positive cancer cells is a more efficient method in conquering cancer than targeting CSCs.
Collapse
Affiliation(s)
- Motoi Fukumoto
- Department of Pathology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Persico M, Petrella L, Orteca N, Di Dato A, Mariani M, Andreoli M, De Donato M, Scambia G, Novellino E, Ferlini C, Fattorusso C. GTP is an allosteric modulator of the interaction between the guanylate-binding protein 1 and the prosurvival kinase PIM1. Eur J Med Chem 2014; 91:132-44. [PMID: 25081641 DOI: 10.1016/j.ejmech.2014.07.093] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 07/21/2014] [Accepted: 07/25/2014] [Indexed: 12/31/2022]
Abstract
GBP1 and PIM1 are known to interact with a molar ratio 1:1. GBP1:PIM1 binding initiates a signaling pathway that induces resistance to common chemotherapeutics such as paclitaxel. Since GBP1 is a large GTPase which undergoes conformational changes in a nucleotide-dependent manner, we investigated the effect of GTP/GDP binding on GBP1:PIM1 interaction by using computational and biological studies. It resulted that only GTP decreases the formation of the GBP1:PIM1 complex through an allosteric mechanism, putting the bases for the identification of new compounds potentially able to revert resistance to paclitaxel.
Collapse
Affiliation(s)
- Marco Persico
- Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, I-80131 Napoli, Italy
| | - Lella Petrella
- Laboratory of Molecular Oncology, Jean Paul II Research Foundation, Campobasso 86100, Italy
| | - Nausicaa Orteca
- Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, I-80131 Napoli, Italy
| | - Antonio Di Dato
- Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, I-80131 Napoli, Italy
| | - Marisa Mariani
- Danbury Hospital Research Institute, Danbury, CT 06810, USA
| | - Mirko Andreoli
- Danbury Hospital Research Institute, Danbury, CT 06810, USA
| | - Marta De Donato
- Catholic University of the Sacred Heart, Department of Obstetrics and Gynaecology, Rome, Italy
| | - Giovanni Scambia
- Catholic University of the Sacred Heart, Department of Obstetrics and Gynaecology, Rome, Italy
| | - Ettore Novellino
- Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, I-80131 Napoli, Italy
| | | | - Caterina Fattorusso
- Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, I-80131 Napoli, Italy.
| |
Collapse
|
42
|
Parker AL, Kavallaris M, McCarroll JA. Microtubules and their role in cellular stress in cancer. Front Oncol 2014; 4:153. [PMID: 24995158 PMCID: PMC4061531 DOI: 10.3389/fonc.2014.00153] [Citation(s) in RCA: 299] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/03/2014] [Indexed: 01/08/2023] Open
Abstract
Microtubules are highly dynamic structures, which consist of α- and β-tubulin heterodimers, and are involved in cell movement, intracellular trafficking, and mitosis. In the context of cancer, the tubulin family of proteins is recognized as the target of the tubulin-binding chemotherapeutics, which suppress the dynamics of the mitotic spindle to cause mitotic arrest and cell death. Importantly, changes in microtubule stability and the expression of different tubulin isotypes as well as altered post-translational modifications have been reported for a range of cancers. These changes have been correlated with poor prognosis and chemotherapy resistance in solid and hematological cancers. However, the mechanisms underlying these observations have remained poorly understood. Emerging evidence suggests that tubulins and microtubule-associated proteins may play a role in a range of cellular stress responses, thus conferring survival advantage to cancer cells. This review will focus on the importance of the microtubule-protein network in regulating critical cellular processes in response to stress. Understanding the role of microtubules in this context may offer novel therapeutic approaches for the treatment of cancer.
Collapse
Affiliation(s)
- Amelia L Parker
- Tumour Biology and Targeting Program, Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales , Sydney, NSW , Australia
| | - Maria Kavallaris
- Tumour Biology and Targeting Program, Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales , Sydney, NSW , Australia ; Australian Centre for NanoMedicine, University of New South Wales , Sydney, NSW , Australia
| | - Joshua A McCarroll
- Tumour Biology and Targeting Program, Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales , Sydney, NSW , Australia ; Australian Centre for NanoMedicine, University of New South Wales , Sydney, NSW , Australia
| |
Collapse
|
43
|
Sox9 and Hif-2α regulate TUBB3 gene expression and affect ovarian cancer aggressiveness. Gene 2014; 542:173-81. [DOI: 10.1016/j.gene.2014.03.037] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/10/2014] [Accepted: 03/17/2014] [Indexed: 02/07/2023]
|
44
|
Abubaker K, Luwor RB, Zhu H, McNally O, Quinn MA, Burns CJ, Thompson EW, Findlay JK, Ahmed N. Inhibition of the JAK2/STAT3 pathway in ovarian cancer results in the loss of cancer stem cell-like characteristics and a reduced tumor burden. BMC Cancer 2014; 14:317. [PMID: 24886434 PMCID: PMC4025194 DOI: 10.1186/1471-2407-14-317] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 04/23/2014] [Indexed: 12/21/2022] Open
Abstract
Background Current treatment of ovarian cancer patients with chemotherapy leaves behind a residual tumor which results in recurrent ovarian cancer within a short time frame. We have previously demonstrated that a single short-term treatment of ovarian cancer cells with chemotherapy in vitro resulted in a cancer stem cell (CSC)-like enriched residual population which generated significantly greater tumor burden compared to the tumor burden generated by control untreated cells. In this report we looked at the mechanisms of the enrichment of CSC-like residual cells in response to paclitaxel treatment. Methods The mechanism of survival of paclitaxel-treated residual cells at a growth inhibitory concentration of 50% (GI50) was determined on isolated tumor cells from the ascites of recurrent ovarian cancer patients and HEY ovarian cancer cell line by in vitro assays and in a mouse xenograft model. Results Treatment of isolated tumor cells from the ascites of ovarian cancer patients and HEY ovarian cancer cell line with paclitaxel resulted in a CSC-like residual population which coincided with the activation of Janus activated kinase 2 (JAK2) and signal transducer and activation of transcription 3 (STAT3) pathway in paclitaxel surviving cells. Both paclitaxel-induced JAK2/STAT3 activation and CSC-like characteristics were inhibited by a low dose JAK2-specific small molecule inhibitor CYT387 (1 μM) in vitro. Subsequent, in vivo transplantation of paclitaxel and CYT387-treated HEY cells in mice resulted in a significantly reduced tumor burden compared to that seen with paclitaxel only-treated transplanted cells. In vitro analysis of tumor xenografts at protein and mRNA levels demonstrated a loss of CSC-like markers and CA125 expression in paclitaxel and CYT387-treated cell-derived xenografts, compared to paclitaxel only-treated cell-derived xenografts. These results were consistent with significantly reduced activation of JAK2 and STAT3 in paclitaxel and CYT387-treated cell-derived xenografts compared to paclitaxel only-treated cell derived xenografts. Conclusions This proof of principle study demonstrates that inhibition of the JAK2/STAT3 pathway by the addition of CYT387 suppresses the ‘stemness’ profile in chemotherapy-treated residual cells in vitro, which is replicated in vivo, leading to a reduced tumor burden. These findings have important implications for ovarian cancer patients who are treated with taxane and/or platinum-based therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Nuzhat Ahmed
- Women's Cancer Research Centre, Royal Women's Hospital, 20 Flemington Road, Parkville, Melbourne, Victoria 3052, Australia.
| |
Collapse
|
45
|
English DP, Roque DM, Santin AD. Class III b-tubulin overexpression in gynecologic tumors: implications for the choice of microtubule targeted agents? Expert Rev Anticancer Ther 2014; 13:63-74. [DOI: 10.1586/era.12.158] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
46
|
Ostler N, Britzen-Laurent N, Liebl A, Naschberger E, Lochnit G, Ostler M, Forster F, Kunzelmann P, Ince S, Supper V, Praefcke GJK, Schubert DW, Stockinger H, Herrmann C, Stürzl M. Gamma interferon-induced guanylate binding protein 1 is a novel actin cytoskeleton remodeling factor. Mol Cell Biol 2014; 34:196-209. [PMID: 24190970 PMCID: PMC3911287 DOI: 10.1128/mcb.00664-13] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/22/2013] [Accepted: 10/28/2013] [Indexed: 02/07/2023] Open
Abstract
Gamma interferon (IFN-γ) regulates immune defenses against viruses, intracellular pathogens, and tumors by modulating cell proliferation, migration, invasion, and vesicle trafficking processes. The large GTPase guanylate binding protein 1 (GBP-1) is among the cellular proteins that is the most abundantly induced by IFN-γ and mediates its cell biologic effects. As yet, the molecular mechanisms of action of GBP-1 remain unknown. Applying an interaction proteomics approach, we identified actin as a strong and specific binding partner of GBP-1. Furthermore, GBP-1 colocalized with actin at the subcellular level and was both necessary and sufficient for the extensive remodeling of the fibrous actin structure observed in IFN-γ-exposed cells. These effects were dependent on the oligomerization and the GTPase activity of GBP-1. Purified GBP-1 and actin bound to each other, and this interaction was sufficient to impair the formation of actin filaments in vitro, as demonstrated by atomic force microscopy, dynamic light scattering, and fluorescence-monitored polymerization. Cosedimentation and band shift analyses demonstrated that GBP-1 binds robustly to globular actin and slightly to filamentous actin. This indicated that GBP-1 may induce actin remodeling via globular actin sequestering and/or filament capping. These results establish GBP-1 as a novel member within the family of actin-remodeling proteins specifically mediating IFN-γ-dependent defense strategies.
Collapse
Affiliation(s)
- Nicole Ostler
- Division of Molecular and Experimental Surgery, University Medical Center Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Nathalie Britzen-Laurent
- Division of Molecular and Experimental Surgery, University Medical Center Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Andrea Liebl
- Division of Molecular and Experimental Surgery, University Medical Center Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, University Medical Center Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Günter Lochnit
- Faculty of Medicine, Institute of Biochemistry, Justus Liebig University, Giessen, Germany
| | - Markus Ostler
- Institute of Physics, Chemnitz University of Technology, Chemnitz, Germany
| | - Florian Forster
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Kunzelmann
- Institute of Polymer Materials, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Semra Ince
- Physical Chemistry I, Ruhr University Bochum, Bochum, Germany
| | - Verena Supper
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Dirk W. Schubert
- Institute of Polymer Materials, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Hannes Stockinger
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Michael Stürzl
- Division of Molecular and Experimental Surgery, University Medical Center Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
47
|
Forster F, Paster W, Supper V, Schatzlmaier P, Sunzenauer S, Ostler N, Saliba A, Eckerstorfer P, Britzen-Laurent N, Schütz G, Schmid JA, Zlabinger GJ, Naschberger E, Stürzl M, Stockinger H. Guanylate binding protein 1-mediated interaction of T cell antigen receptor signaling with the cytoskeleton. THE JOURNAL OF IMMUNOLOGY 2013; 192:771-81. [PMID: 24337748 DOI: 10.4049/jimmunol.1300377] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
GTPases act as important switches in many signaling events in cells. Although small and heterotrimeric G proteins are subjects of intensive studies, little is known about the large IFN-inducible GTPases. In this article, we show that the IFN-γ-inducible guanylate binding protein 1 (GBP-1) is a regulator of T cell activation. Silencing of GBP-1 leads to enhanced activation of early T cell Ag receptor/CD3 signaling molecules, including Lck, that is translated to higher IL-2 production. Mass spectrometry analyses showed that regulatory cytoskeletal proteins, like plastin-2 that bundles actin fibers and spectrin β-chain, brain 1 that links the plasma membrane to the actin cytoskeleton, are binding partners of GBP-1. The spectrin cytoskeleton influences cell spreading and surface expression of TCR/CD3 and the leukocyte phosphatase CD45. We found higher cell spreading and enhanced surface expression of TCR/CD3 and CD45 in GBP-1 silenced T cells that explain their enhanced TCR/CD3 signaling. We conclude that GBP-1 is a downstream processor of IFN-γ via which T cells regulate cytoskeleton-dependent cell functions.
Collapse
Affiliation(s)
- Florian Forster
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Roque DM, Buza N, Glasgow M, Bellone S, Bortolomai I, Gasparrini S, Cocco E, Ratner E, Silasi DA, Azodi M, Rutherford TJ, Schwartz PE, Santin AD. Class III β-tubulin overexpression within the tumor microenvironment is a prognostic biomarker for poor overall survival in ovarian cancer patients treated with neoadjuvant carboplatin/paclitaxel. Clin Exp Metastasis 2013; 31:101-10. [PMID: 24005572 DOI: 10.1007/s10585-013-9614-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 08/11/2013] [Indexed: 11/26/2022]
Abstract
Critics have suggested that neoadjuvant chemotherapy (NACT) followed by interval debulking may select for resistant clones or cancer stem cells when compared to primary cytoreduction. β-tubulins are chemotherapeutic targets of taxanes and epothilones. Class III β-tubulin overexpression has been linked to chemoresistance and hypoxia. Herein, we describe changes in class III β-tubulin in patients with advanced ovarian carcinoma in response to NACT, in relationship to clinical outcome, and between patients who underwent NACT versus primary debulking; we characterize in vitro chemosensitivity to paclitaxel/patupilone of cell lines established from this patient population, and class III β-tubulin expression following repeated exposure to paclitaxel. Using immunohistochemistry, we observed among 22 paired specimens obtained before/after NACT decreased expression of class III β-tubulin following therapy within stroma (p=0.07), but not tumor (p=0.63). Poor median overall survival was predicted by high levels of class III β-tubulin in both tumor (HR 3.66 [1.11,12.05], p=0.03) and stroma (HR 4.53 [1.28,16.1], p=0.02). Class III β-tubulin expression by quantitative-real-time-polymerase-chain-reaction was higher among patients who received NACT (n=12) compared to primary cytoreduction (n=14) (mean±SD fold-change: 491.2±115.9 vs. 224.1±55.66, p=0.037). In vitro subculture with paclitaxel resulted in class III β-tubulin upregulation, however, cell lines that overexpressed class III β-tubulin remained sensitive to patupilone. Overexpression of class III β-tubulin in patients dispositioned to NACT may thus identify an intrinsically aggressive phenotype, and predict poor overall survival and paclitaxel resistance. Decreases in stromal expression may represent normalization of the tumor microenvironment following therapy. Epothilones warrant study for patients who have received neoadjuvant carboplatin and paclitaxel.
Collapse
Affiliation(s)
- Dana M Roque
- Division of Gynecologic Oncology, Yale University School of Medicine, 333 Cedar Street FMB 328, Box 208063, New Haven, CT, 06520, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Mitochondrial (mt) dysfunction in gliomas has been linked to abnormalities of mt energy metabolism, marked by a metabolic shift from oxidative phosphorylation to glycolysis ("Warburg effect"), disturbances in mt membrane potential regulation and apoptotic signaling, as well as to somatic mutations involving the Krebs cycle enzyme isocitrate dehydrogenase. Evolving biological concepts with potential therapeutic implications include interaction between microtubule proteins and mitochondria (mt) in the control of closure of voltage-dependent anion channels and in the regulation of mt dynamics and the mt-endoplasmic reticulum network. The cytoskeletal protein βIII-tubulin, which is overexpressed in malignant gliomas, has emerged as a prosurvival factor associated in part with mt and also as a marker of chemoresistance. Mt-targeted therapeutic strategies that are discussed include the following: (1) metabolic modulation with emphasis on dichloroacetate, a pyruvate dehydrogenase kinase inhibitor; (2) tumor cell death via apoptosis induced by tricyclic antidepressants, microtubule-modulating drugs, and small molecules or compounds capable of inflicting reactive oxygen species-dependent tumor cell death; and (3) pretreatment mt priming and mt-targeted prodrug cancer therapy.
Collapse
|
50
|
Lobert S, Graichen ME, Morris K. Coordinated Regulation of β-Tubulin Isotypes and Epithelial-to-Mesenchymal Transition Protein ZEB1 in Breast Cancer Cells. Biochemistry 2013; 52:5482-90. [DOI: 10.1021/bi400340g] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Sharon Lobert
- School of
Nursing, University of Mississippi Medical Center, 2500 North State Street, Jackson, Mississippi
39216, United States
| | - Mary E. Graichen
- School of
Nursing, University of Mississippi Medical Center, 2500 North State Street, Jackson, Mississippi
39216, United States
| | - Kevin Morris
- School of
Nursing, University of Mississippi Medical Center, 2500 North State Street, Jackson, Mississippi
39216, United States
| |
Collapse
|