1
|
Li DZ, Yang ZY, Leng A, Zhang Q, Zhang XD, Bian YC, Xiao R, Ren JJ. Targeting AGTPBP1 inhibits pancreatic cancer progression via regulating microtubules and ERK signaling pathway. Mol Med 2024; 30:119. [PMID: 39129004 PMCID: PMC11318240 DOI: 10.1186/s10020-024-00892-x] [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: 01/23/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND AGTPBP1 is a cytosolic carboxypeptidase that cleaves poly-glutamic acids from the C terminus or side chains of α/β tubulins. Although its dysregulated expression has been linked to the development of non-small cell lung cancer, the specific roles and mechanisms of AGTPBP1 in pancreatic cancer (PC) have yet to be fully understood. In this study, we examined the role of AGTPBP1 on PC in vitro and in vivo. METHODS Immunohistochemistry was used to examine the expression of AGTPBP1 in PC and non-cancerous tissues. Additionally, we assessed the malignant behaviors of PC cells following siRNA-mediated AGTPBP1 knockdown both in vitro and in vivo. RNA sequencing and bioinformatics analysis were performed to identify the differentially expressed genes regulated by AGTPBP1. RESULTS We determined that AGTPBP1 was overexpressed in PC tissues and the higher expression of AGTPBP1 was closely related to the location of tumors. AGTPBP1 inhibition can significantly decrease cell progression in vivo and in vitro. Moreover, the knockdown of AGTPBP1 inhibited the expression of ERK1/2, P-ERK1/2, MYLK, and TUBB4B proteins via the ERK signaling pathway. CONCLUSION Our research indicates that AGTPBP1 may be a putative therapeutic target for PC.
Collapse
Affiliation(s)
- Ding-Zhong Li
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, #1, Tongdao North Street, Huhhot, 010051, PR China
| | - Zhe-Yu Yang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, #1, Tongdao North Street, Huhhot, 010051, PR China
| | - Asi Leng
- Medical Simulation Center, Inner Mongolia Medical University, Huhhot, 010059, PR China
| | - Qian Zhang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, #1, Tongdao North Street, Huhhot, 010051, PR China
| | - Xiao-Dong Zhang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, #1, Tongdao North Street, Huhhot, 010051, PR China
| | - Yan-Chao Bian
- Inner Mongolia Key Laboratory of Molecular Pathology, Inner Mongolia Medical University, #5, Xin Hua Street, Huhhot, 010059, PR China
| | - Rui Xiao
- Inner Mongolia Key Laboratory of Molecular Pathology, Inner Mongolia Medical University, #5, Xin Hua Street, Huhhot, 010059, PR China.
| | - Jian-Jun Ren
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, #1, Tongdao North Street, Huhhot, 010051, PR China.
| |
Collapse
|
2
|
Amiri A, Dietz C, Rapp A, Cardoso MC, Stark RW. The cyto-linker and scaffolding protein "plectin" mis-localization leads to softening of cancer cells. NANOSCALE 2023; 15:15008-15026. [PMID: 37668423 DOI: 10.1039/d3nr02226a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Discovering tools to prevent cancer progression requires understanding the fundamental differences between normal and cancer cells. More than a decade ago, atomic force microscopy (AFM) revealed cancer cells' softer body compared to their healthy counterparts. Here, we investigated the mechanism underlying the softening of cancerous cells in comparison with their healthy counterparts based on AFM high resolution stiffness tomography and 3D confocal microscopy. We showed microtubules (MTs) network in invasive ductal carcinoma cell cytoskeleton is basally located and segmented for around 400 nm from the cell periphery. Additionally, the cytoskeleton scaffolding protein plectin exhibits a mis-localization from the cytoplasm to the surface of cells in the carcinoma which justifies the dissociation of the MT network from the cell's cortex. Furthermore, the assessment of MTs' persistence length using a worm-like-chain (WLC) model in high resolution AFM images showed lower persistence length of the single MTs in ductal carcinoma compared to that in the normal state. Overall, these tuned mechanics support the invasive cells to ascertain more flexibility under compressive forces in small deformations. These data provide new insights into the structural origins of cancer aids in progression.
Collapse
Affiliation(s)
- Anahid Amiri
- Physics of Surfaces, Institute of Materials Science, Technical University of Darmstadt, Alarich-Weiss-Str. 2, 64287 Darmstadt, Germany.
| | - Christian Dietz
- Physics of Surfaces, Institute of Materials Science, Technical University of Darmstadt, Alarich-Weiss-Str. 2, 64287 Darmstadt, Germany.
| | - Alexander Rapp
- Cell Biology and Epigenetics, Department of Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - M Cristina Cardoso
- Cell Biology and Epigenetics, Department of Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Robert W Stark
- Physics of Surfaces, Institute of Materials Science, Technical University of Darmstadt, Alarich-Weiss-Str. 2, 64287 Darmstadt, Germany.
| |
Collapse
|
3
|
Kumar N, Goel N. Recent development of imidazole derivatives as potential anticancer agents. PHYSICAL SCIENCES REVIEWS 2022. [DOI: 10.1515/psr-2021-0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Abstract
Cancer, one of the key health problems globally, is a group of related diseases that share a number of characteristics primarily the uncontrolled growth and invasive to surrounding tissues. Chemotherapy is one of the ways for the treatment of cancer which uses one or more anticancer agents as per chemotherapy regimen. Limitations of most anticancer drugs due to a variety of reasons such as serious side effects, drug resistance, lack of sensitivity and efficacy etc. generate the necessity towards the designing of novel anticancer lead molecules. In this regard, the synthesis of biologically active heterocyclic molecules is an appealing research area. Among heterocyclic compounds, nitrogen containing heterocyclic molecules has fascinated tremendous consideration due to broad range of pharmaceutical activity. Imidazoles, extensively present in natural products as well as synthetic molecules, have two nitrogen atoms, and are five membered heterocyclic rings. Because of their countless physiological and pharmacological characteristics, medicinal chemists are enthused to design and synthesize new imidazole derivatives with improved pharmacodynamic and pharmacokinetic properties. The aim of this present chapter is to discuss the synthesis, chemistry, pharmacological activity, and scope of imidazole-based molecules in anticancer drug development. Finally, we have discussed the current challenges and future perspectives of imidazole-based derivatives in anticancer drug development.
Collapse
Affiliation(s)
- Naresh Kumar
- Department of Biosciences and Biomedical Engineering , Indian Institute of Technology Indore , Indore , Madhya Pradesh 453552 , India
| | - Nidhi Goel
- Department of Chemistry , Institute of Science, Banaras Hindu University , Varanasi , Uttar Pradesh 221005 , India
| |
Collapse
|
4
|
Principe DR, Underwood PW, Korc M, Trevino JG, Munshi HG, Rana A. The Current Treatment Paradigm for Pancreatic Ductal Adenocarcinoma and Barriers to Therapeutic Efficacy. Front Oncol 2021; 11:688377. [PMID: 34336673 PMCID: PMC8319847 DOI: 10.3389/fonc.2021.688377] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, with a median survival time of 10-12 months. Clinically, these poor outcomes are attributed to several factors, including late stage at the time of diagnosis impeding resectability, as well as multi-drug resistance. Despite the high prevalence of drug-resistant phenotypes, nearly all patients are offered chemotherapy leading to modest improvements in postoperative survival. However, chemotherapy is all too often associated with toxicity, and many patients elect for palliative care. In cases of inoperable disease, cytotoxic therapies are less efficacious but still carry the same risk of serious adverse effects, and clinical outcomes remain particularly poor. Here we discuss the current state of pancreatic cancer therapy, both surgical and medical, and emerging factors limiting the efficacy of both. Combined, this review highlights an unmet clinical need to improve our understanding of the mechanisms underlying the poor therapeutic responses seen in patients with PDAC, in hopes of increasing drug efficacy, extending patient survival, and improving quality of life.
Collapse
Affiliation(s)
- Daniel R. Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, United States
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Murray Korc
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Jose G. Trevino
- Department of Surgery, Division of Surgical Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - Hidayatullah G. Munshi
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Jesse Brown VA Medical Center, Chicago, IL, United States
| | - Ajay Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
- Jesse Brown VA Medical Center, Chicago, IL, United States
| |
Collapse
|
5
|
Sharbeen G, McCarroll JA, Akerman A, Kopecky C, Youkhana J, Kokkinos J, Holst J, Boyer C, Erkan M, Goldstein D, Timpson P, Cox TR, Pereira BA, Chitty JL, Fey SK, Najumudeen AK, Campbell AD, Sansom OJ, Ignacio RMC, Naim S, Liu J, Russia N, Lee J, Chou A, Johns A, Gill AJ, Gonzales-Aloy E, Gebski V, Guan YF, Pajic M, Turner N, Apte MV, Davis TP, Morton JP, Haghighi KS, Kasparian J, McLean BJ, Setargew YF, Phillips PA. Cancer-Associated Fibroblasts in Pancreatic Ductal Adenocarcinoma Determine Response to SLC7A11 Inhibition. Cancer Res 2021; 81:3461-3479. [PMID: 33980655 DOI: 10.1158/0008-5472.can-20-2496] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 03/01/2021] [Accepted: 04/28/2021] [Indexed: 11/16/2022]
Abstract
Cancer-associated fibroblasts (CAF) are major contributors to pancreatic ductal adenocarcinoma (PDAC) progression through protumor signaling and the generation of fibrosis, the latter of which creates a physical barrier to drugs. CAF inhibition is thus an ideal component of any therapeutic approach for PDAC. SLC7A11 is a cystine transporter that has been identified as a potential therapeutic target in PDAC cells. However, no prior study has evaluated the role of SLC7A11 in PDAC tumor stroma and its prognostic significance. Here we show that high expression of SLC7A11 in human PDAC tumor stroma, but not tumor cells, is independently prognostic of poorer overall survival. Orthogonal approaches showed that PDAC-derived CAFs are highly dependent on SLC7A11 for cystine uptake and glutathione synthesis and that SLC7A11 inhibition significantly decreases CAF proliferation, reduces their resistance to oxidative stress, and inhibits their ability to remodel collagen and support PDAC cell growth. Importantly, specific ablation of SLC7A11 from the tumor compartment of transgenic mouse PDAC tumors did not affect tumor growth, suggesting the stroma can substantially influence PDAC tumor response to SLC7A11 inhibition. In a mouse orthotopic PDAC model utilizing human PDAC cells and CAFs, stable knockdown of SLC7A11 was required in both cell types to reduce tumor growth, metastatic spread, and intratumoral fibrosis, demonstrating the importance of targeting SLC7A11 in both compartments. Finally, treatment with a nanoparticle gene-silencing drug against SLC7A11, developed by our laboratory, reduced PDAC tumor growth, incidence of metastases, CAF activation, and fibrosis in orthotopic PDAC tumors. Overall, these findings identify an important role of SLC7A11 in PDAC-derived CAFs in supporting tumor growth. SIGNIFICANCE: This study demonstrates that SLC7A11 in PDAC stromal cells is important for the tumor-promoting activity of CAFs and validates a clinically translatable nanomedicine for therapeutic SLC7A11 inhibition in PDAC.
Collapse
Affiliation(s)
- George Sharbeen
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Joshua A McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales Sydney, New South Wales, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, New South Wales, Australia
| | - Anouschka Akerman
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Chantal Kopecky
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Janet Youkhana
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - John Kokkinos
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales Sydney, New South Wales, Australia
| | - Jeff Holst
- School of Medical Science and Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Cyrille Boyer
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales Sydney, New South Wales, Australia
| | - Mert Erkan
- Koc University Research Centre for Translational Medicine and Department of Surgery, Koc University, School of Medicine, Istanbul, Turkey
| | - David Goldstein
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
- Prince of Wales Hospital, Prince of Wales Clinical School, Sydney, New South Wales, Australia
| | - Paul Timpson
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Thomas R Cox
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Brooke A Pereira
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Jessica L Chitty
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Sigrid K Fey
- Cancer Research UK, Beatson Institute, Glasgow, United Kingdom
| | | | | | - Owen J Sansom
- Cancer Research UK, Beatson Institute, Glasgow, United Kingdom
| | - Rosa Mistica C Ignacio
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Stephanie Naim
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Jie Liu
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Nelson Russia
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Julia Lee
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Angela Chou
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Department of Anatomical Pathology, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Amber Johns
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
| | - Anthony J Gill
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Estrella Gonzales-Aloy
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Val Gebski
- NHMRC Clinical Trials Centre, University of Sydney, New South Wales, Australia
| | - Yi Fang Guan
- School of Medical Science and Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Marina Pajic
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
| | - Nigel Turner
- School of Medical Sciences, University of New South Wales Sydney, New South Wales, Australia
| | - Minoti V Apte
- Pancreatic Research Group, South Western Sydney Clinical School, University New South Wales and Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Institute of Bioengineering & Nanotechnology, University of Queensland, Queensland, Australia
| | - Jennifer P Morton
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Koroush S Haghighi
- Prince of Wales Hospital, Prince of Wales Clinical School, Sydney, New South Wales, Australia
| | - Jorjina Kasparian
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Benjamin J McLean
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | | | - Phoebe A Phillips
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia.
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Albahde MAH, Abdrakhimov B, Li GQ, Zhou X, Zhou D, Xu H, Qian H, Wang W. The Role of Microtubules in Pancreatic Cancer: Therapeutic Progress. Front Oncol 2021; 11:640863. [PMID: 34094924 PMCID: PMC8176010 DOI: 10.3389/fonc.2021.640863] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer has an extremely low prognosis, which is attributable to its high aggressiveness, invasiveness, late diagnosis, and lack of effective therapies. Among all the drugs joining the fight against this type of cancer, microtubule-targeting agents are considered to be the most promising. They inhibit cancer cells although through different mechanisms such as blocking cell division, apoptosis induction, etc. Hereby, we review the functions of microtubule cytoskeletal proteins in tumor cells and comprehensively examine the effects of microtubule-targeting agents on pancreatic carcinoma.
Collapse
Affiliation(s)
- Mugahed Abdullah Hasan Albahde
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
| | - Bulat Abdrakhimov
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guo-Qi Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Xiaohu Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Dongkai Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Hao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Huixiao Qian
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
7
|
Boyle ST, Mittal P, Kaur G, Hoffmann P, Samuel MS, Klingler-Hoffmann M. Uncovering Tumor-Stroma Inter-relationships Using MALDI Mass Spectrometry Imaging. J Proteome Res 2020; 19:4093-4103. [PMID: 32870688 DOI: 10.1021/acs.jproteome.0c00511] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumorigenesis involves a complex interplay between genetically modified cancer cells and their adjacent normal tissue, the stroma. We used an established breast cancer mouse model to investigate this inter-relationship. Conditional activation of Rho-associated protein kinase (ROCK) in a model of mammary tumorigenesis enhances tumor growth and progression by educating the stroma and enhancing the production and remodeling of the extracellular matrix. We used peptide matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to quantify the proteomic changes occurring within tumors and their stroma in their regular spatial context. Peptides were ranked according to their ability to discriminate between the two groups, using a receiver operating characteristic tool. Peptides were identified by liquid chromatography tandem mass spectrometry, and protein expression was validated by quantitative immunofluorescence using an independent set of tumor samples. We have identified and validated four key proteins upregulated in ROCK-activated mammary tumors relative to those expressing kinase-dead ROCK, namely, collagen I, α-SMA, Rab14, and tubulin-β4. Rab14 and tubulin-β4 are expressed within tumor cells, whereas collagen I is localized within the stroma. α-SMA is predominantly localized within the stroma but is also expressed at higher levels in the epithelia of ROCK-activated tumors. High expression of COL1A, the gene encoding the pro-α 1 chain of collagen, correlates with cancer progression in two human breast cancer genomic data sets, and high expression of COL1A and ACTA2 (the gene encoding α-SMA) are associated with a low survival probability (COLIA, p = 0.00013; ACTA2, p = 0.0076) in estrogen receptor-negative breast cancer patients. To investigate whether ROCK-activated tumor cells cause stromal cancer-associated fibroblasts (CAFs) to upregulate expression of collagen I and α-SMA, we treated CAFs with medium conditioned by primary mammary tumor cells in which ROCK had been activated. This led to abundant production of both proteins in CAFs, clearly highlighting the inter-relationship between tumor cells and CAFs and identifying CAFs as the potential source of high levels of collagen 1 and α-SMA and associated enhancement of tissue stiffness. Our research emphasizes the capacity of MALDI-MSI to quantitatively assess tumor-stroma inter-relationships and to identify potential prognostic factors for cancer progression in human patients, using sophisticated mouse cancer models.
Collapse
Affiliation(s)
- Sarah T Boyle
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide SA 5000, Australia
| | - Parul Mittal
- Future Industries Institute, University of South Australia, Mawson Lakes SA 5095, Australia
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden 11800 Pulau Pinang, Malaysia
| | - Peter Hoffmann
- Future Industries Institute, University of South Australia, Mawson Lakes SA 5095, Australia
| | - Michael S Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide SA 5000, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide SA 5000, Australia
| | | |
Collapse
|
8
|
Borys F, Joachimiak E, Krawczyk H, Fabczak H. Intrinsic and Extrinsic Factors Affecting Microtubule Dynamics in Normal and Cancer Cells. Molecules 2020; 25:E3705. [PMID: 32823874 PMCID: PMC7464520 DOI: 10.3390/molecules25163705] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/18/2022] Open
Abstract
Microtubules (MTs), highly dynamic structures composed of α- and β-tubulin heterodimers, are involved in cell movement and intracellular traffic and are essential for cell division. Within the cell, MTs are not uniform as they can be composed of different tubulin isotypes that are post-translationally modified and interact with different microtubule-associated proteins (MAPs). These diverse intrinsic factors influence the dynamics of MTs. Extrinsic factors such as microtubule-targeting agents (MTAs) can also affect MT dynamics. MTAs can be divided into two main categories: microtubule-stabilizing agents (MSAs) and microtubule-destabilizing agents (MDAs). Thus, the MT skeleton is an important target for anticancer therapy. This review discusses factors that determine the microtubule dynamics in normal and cancer cells and describes microtubule-MTA interactions, highlighting the importance of tubulin isoform diversity and post-translational modifications in MTA responses and the consequences of such a phenomenon, including drug resistance development.
Collapse
Affiliation(s)
- Filip Borys
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, 00-664 Warsaw, Poland;
| | - Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
| | - Hanna Krawczyk
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, 00-664 Warsaw, Poland;
| | - Hanna Fabczak
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
| |
Collapse
|
9
|
Kamal MA, Al-Zahrani MH, Khan SH, Khan MH, Al-Subhi HA, Kuerban A, Aslam M, Al-Abbasi FA, Anwar F. Tubulin Proteins in Cancer Resistance: A Review. Curr Drug Metab 2020; 21:178-185. [DOI: 10.2174/1389200221666200226123638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 12/26/2022]
Abstract
Cancer cells are altered with cell cycle genes or they are mutated, leading to a high rate of proliferation
compared to normal cells. Alteration in these genes leads to mitosis dysregulation and becomes the basis of tumor
progression and resistance to many drugs. The drugs which act on the cell cycle fail to arrest the process, making
cancer cell non-responsive to apoptosis or cell death. Vinca alkaloids and taxanes fall in this category and are
referred to as antimitotic agents. Microtubule proteins play an important role in mitosis during cell division as a
target site for vinca alkaloids and taxanes. These proteins are dynamic in nature and are composed of α-β-tubulin
heterodimers. β-tubulin specially βΙΙΙ isotype is generally altered in expression within cancerous cells. Initially,
these drugs were very effective in the treatment of cancer but failed to show their desired action after initial
chemotherapy. The present review highlights some of the important targets and their mechanism of resistance
offered by cancer cells with new promising drugs from natural sources that can lead to the development of a new
approach to chemotherapy.
Collapse
Affiliation(s)
- Mohammad Amjad Kamal
- Metabolomics and Enzymology Unit, Fundamental and Applied Biology Group, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Maryam Hassan Al-Zahrani
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Salman Hasan Khan
- Department of Orthodontics, and Dentofacial Orthopaedics, TMU Dental College, Moradabad, Uttar Pradesh, India
| | - Mateen Hasan Khan
- Department of Pharmacology, Shri Venkateshwara University, Gajraula, Amroha, Uttar Pradesh, India
| | - Hani Awad Al-Subhi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abudukadeer Kuerban
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Muhammad Aslam
- Department of Statistics, Faculy of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Fahad Ahmed Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Firoz Anwar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
10
|
Albahde MAH, Zhang P, Zhang Q, Li G, Wang W. Upregulated Expression of TUBA1C Predicts Poor Prognosis and Promotes Oncogenesis in Pancreatic Ductal Adenocarcinoma via Regulating the Cell Cycle. Front Oncol 2020; 10:49. [PMID: 32117719 PMCID: PMC7033491 DOI: 10.3389/fonc.2020.00049] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant disease and has the worst prognosis and survival rate. TUBA1C is a microtubule component implicated in multiple cancers, however, the clinical significance and biological functions of TUBA1C in the progression of PDAC remain unexplored. Methods: The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis (GEPIA) data were employed to detect the TUBA1C mRNA expression and the relation between TUBA1C expression and overall survival (OS) in PDAC. Then, bioinformatic analysis was employed to determine the potential pathway and genes related to TUBA1C. Human pancreatic cancer tissue and adjacent non-tumor tissues samples were detected by immunochemistry (IHC) staining, and the correlation between TUBA1C expression and the clinicopathological features were investigated. Meanwhile, TUBA1C expression in PDAC cell lines was evaluated by western blotting. Furthermore, functional assays including cell viability, apoptosis, cell cycle, transwell assay, wound healing assay, and a xenograft tumor model were performed to determine the oncogenic role of TUBA1C in PDAC, respectively. Results: TUBA1C was overexpressed in the PDAC tissues and cells. IHC analysis showed that the TUBA1C overexpression was associated with short OS. Bioinformatic analysis indicated that TUBA1C overexpression was mainly associated with cell cycle regulation. The downregulation of TUBA1C significantly suppressed cell proliferation, induced cell apoptosis and cycle arrest, and inhibited invasion and migration in PDAC cells. Furthermore, TUBA1C downregulation also inhibited tumor growth in vivo. Conclusion: These findings suggested that TUBA1C downregulation suppressed PDAC aggressiveness via cell cycle pathway and that TUBA1C may serve as a potential prognostic marker for PDAC therapy.
Collapse
Affiliation(s)
- Mugahed Abdullah Hasan Albahde
- Department of Hepatobiliary and Pancreatic Surgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China.,Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China.,Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, China
| | - Piao Zhang
- Department of Anesthesiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Qiuqiang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Guoqi Li
- Department of Hepatobiliary and Pancreatic Surgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China.,Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China.,Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, China.,Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
11
|
Li L, Quan D, Chen J, Ding J, Zhao J, Lv L, Chen J. Design, synthesis, and biological evaluation of 1-substituted -2-aryl imidazoles targeting tubulin polymerization as potential anticancer agents. Eur J Med Chem 2019; 184:111732. [PMID: 31610372 DOI: 10.1016/j.ejmech.2019.111732] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/12/2019] [Accepted: 09/21/2019] [Indexed: 01/21/2023]
Abstract
A series of novel 1-substituted-2-aryl imidazoles (SAI) were designed and synthesized based on our previously reported ABI (2-Aryl-4-Benzoyl Imidazole) analogs and on the structure of combretastatin A-4 (CA-4). These compounds showed potent antiproliferative activities against six human cancer cell lines with IC50 values in nano molar range. Among them, compound 3X exhibited the best anticancer activity with an average IC50 value of ∼100 nM. The compound maintains the mechanism of action by inhibiting tubulin polymerization, thus causing cell arrest at G2/M phase and apoptosis. In vivo efficacy studies indicated that 3X was highly effective in suppressing tumor growth in a MDA-MB-468 xenograft model of nude mouse with a TGI (Tumor Growth Suppression) of 77% at 60 mg/kg without causing significant toxicity. In addition, 3X displayed significantly better water solubility (36.70 μg/mL) than CA-4 (2.83 μg/mL). Molecular modeling study indicated that 3X binds well to the colchicine binding site in tubulin. Our results suggest that the novel SAI analogs deserve further investigation as potential anticancer agents.
Collapse
Affiliation(s)
- Ling Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Dongling Quan
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jingxuan Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiahao Ding
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jinwu Zhao
- School of Pharmacy, Guangdong Medical University, Songshan Lake Science and Technology Industry Park, Dongguan, 523808, China
| | - Lin Lv
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Kashyap VK, Wang Q, Setua S, Nagesh PKB, Chauhan N, Kumari S, Chowdhury P, Miller DD, Yallapu MM, Li W, Jaggi M, Hafeez BB, Chauhan SC. Therapeutic efficacy of a novel βIII/βIV-tubulin inhibitor (VERU-111) in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:29. [PMID: 30674344 PMCID: PMC6343279 DOI: 10.1186/s13046-018-1009-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
Abstract
Background The management of pancreatic cancer (PanCa) is exceptionally difficult due to poor response to available therapeutic modalities. Tubulins play a major role in cell dynamics, thus are important molecular targets for cancer therapy. Among various tubulins, βIII and βIV-tubulin isoforms have been primarily implicated in PanCa progression, metastasis and chemo-resistance. However, specific inhibitors of these isoforms that have potent anti-cancer activity with low toxicity are not readily available. Methods We determined anti-cancer molecular mechanisms and therapeutic efficacy of a novel small molecule inhibitor (VERU-111) using in vitro (MTS, wound healing, Boyden chamber and real-time xCELLigence assays) and in vivo (xenograft studies) models of PanCa. The effects of VERU-111 treatment on the expression of β-tubulin isoforms, apoptosis, cancer markers and microRNAs were determined by Western blot, immunohistochemistry (IHC), confocal microscopy, qRT-PCR and in situ hybridization (ISH) analyses. Results We have identified a novel small molecule inhibitor (VERU-111), which preferentially represses clinically important, βIII and βIV tubulin isoforms via restoring the expression of miR-200c. As a result, VERU-111 efficiently inhibited tumorigenic and metastatic characteristics of PanCa cells. VERU-111 arrested the cell cycle in the G2/M phase and induced apoptosis in PanCa cell lines via modulation of cell cycle regulatory (Cdc2, Cdc25c, and Cyclin B1) and apoptosis - associated (Bax, Bad, Bcl-2, and Bcl-xl) proteins. VERU-111 treatment also inhibited tumor growth (P < 0.01) in a PanCa xenograft mouse model. Conclusions This study has identified an inhibitor of βIII/βIV tubulins, which appears to have excellent potential as monotherapy or in combination with conventional therapeutic regimens for PanCa treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-1009-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vivek K Kashyap
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Qinghui Wang
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Saini Setua
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Prashanth K B Nagesh
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Neeraj Chauhan
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Sonam Kumari
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| | - Bilal Bin Hafeez
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
13
|
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
|