1
|
Yang Z, Mogre S, Jun H, He R, Ghosh Chaudhary S, Bhattarai UR, Ho Sui SJ, Matulonis UA, Lazo S, Shetty A, Cameron A, Nguyen QD, Hill SJ. Functional Profiling of p53 and RB Cell Cycle Regulatory Proficiency Suggests Mechanism-Driven Molecular Stratification in Endometrial Carcinoma. CANCER RESEARCH COMMUNICATIONS 2025; 5:719-742. [PMID: 39932274 PMCID: PMC12042793 DOI: 10.1158/2767-9764.crc-24-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/25/2024] [Accepted: 02/07/2025] [Indexed: 02/19/2025]
Abstract
SIGNIFICANCE We show novel cell cycle regulatory molecular classifications and therapeutic targets for endometrial carcinoma. Intact RB regulation and mitotic progression regulatory defects correlate with CDK4/6 and Aurora kinase B inhibitor sensitivity respectively.
Collapse
Affiliation(s)
- Zelei Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Saie Mogre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hyeji Jun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ruiyang He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sneha Ghosh Chaudhary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Upendra Raj Bhattarai
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Shannan J. Ho Sui
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Ursula A. Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Suzan Lazo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Aniket Shetty
- Center for Patient-Derived Models, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amy Cameron
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Quang-Dé Nguyen
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sarah J. Hill
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
2
|
Wang C, Liu J, Wu Y, Cai C, Chai Z, Jia P, Yuan Y, Jiang Z. AURKB as a Therapeutic Target and Key Driver of Liver Cancer Growth and Metastasis. APMIS 2025; 133:e70021. [PMID: 40177797 DOI: 10.1111/apm.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/05/2025]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related deaths worldwide. Aurora kinase B (AURKB), a critical regulator of mitosis, has been implicated in cancer progression, though its precise role in HCC remains unclear. In this study, AURKB expression was found to be significantly elevated in HCC tissues and cell lines compared to controls, as validated by GEPIA and ENCORI databases. Functional assays revealed that AURKB knockdown reduced cell proliferation, invasion, and migration, while increasing apoptosis. Furthermore, suppression of AURKB affected epithelial-mesenchymal transition (EMT) markers, decreasing vimentin and N-cadherin levels and increasing E-cadherin expression. In vivo, a xenograft mouse model demonstrated that tumors derived from AURKB-silenced cells exhibited reduced growth and fewer lung metastases. Histological and immunohistochemical analyses showed lower levels of Ki-67, MMP-9, and EMT markers in these tumors, alongside increased E-cadherin. These findings highlight AURKB's critical role in promoting HCC progression, metastasis, and EMT regulation. Overexpression of AURKB was associated with poor prognosis, suggesting it could serve as a potential biomarker and therapeutic target for liver cancer. Overall, targeting AURKB may provide a novel approach to inhibit HCC growth and metastasis, improving patient outcomes.
Collapse
Affiliation(s)
- Chen Wang
- Department of Interventional Vascular, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Jiangwen Liu
- Department of Hepatobilary Surgery, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Yali Wu
- Department of Insurance Office, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Chen Cai
- Department of Interventional Vascular, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Zhiwei Chai
- Department of Medical, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Ping Jia
- Department of Catheterization Room, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Yueyue Yuan
- Department of Interventional Vascular, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Zhixin Jiang
- Department of Hepatobilary Surgery, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
3
|
Gupta D, Shukla PK, Chowdhury S, Kumari S, Kaur P, Kumar M. "Exploration of Novel Anticancerous Agents Targeting Human Aurora Kinase C". J Cell Biochem 2025; 126:e70025. [PMID: 40123311 DOI: 10.1002/jcb.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
Aurora kinases (AKs), a family of serine/threonine kinases, play a vital role in chromosome segregation during the cell cycle (Mountzios et al., 2008). This family includes Aurora Kinase A (AKA), Aurora Kinase B (AKB), and Aurora Kinase C (AKC). AKA and AKB are active during mitosis, while AKC is involved mostly in germ cell as well as somatic cells. Elevated levels of AKC have been found in several cancer cell lines including breast, cervical, thyroid, colorectal, and liver cancers, making it a significant target for cancer therapy (Tang et al., 2017). In cancers such as glioblastoma and prostate cancer, for example, AKC up regulation has been associated with increased tumor aggressiveness, highlighting its potential role in tumor progression and poor prognosis. Our study employs computational methods, including molecular docking and structure-based virtual screening, to explore a data set of 2 65 241 compounds from the National Cancer Institute (NCI) database, focusing on AKC as a potential target for drug discovery. Through docking studies, several promising compounds that interact with the enzyme's ATP binding pocket, particularly with residues Phe54, Lys72, Ala123, Glu121 and Glu127 of AKC, were identified. The stability of these interactions was assessed through 200-ns molecular dynamics (MD) simulations, revealing that the majority of compounds exhibited stable interactions, while a few displayed fluctuations in their trajectories. Most compounds adhered to favorable pharmacokinetic properties. Comprehensive MD simulations and free energy calculations identified three top candidates (90 729, 37 623, and 134 546) with strong potential as potent inhibitors of AKC. Additional in vitro and in vivo studies are required to confirm the therapeutic potential of these candidates.
Collapse
Affiliation(s)
- Deepali Gupta
- Department of Biophysics, All India Institute of Medical Sciences, Delhi, India
| | - Prakash Kumar Shukla
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, India
| | | | - Supriya Kumari
- Department of Biophysics, All India Institute of Medical Sciences, Delhi, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, Delhi, India
| | - Mukesh Kumar
- Department of Biophysics, All India Institute of Medical Sciences, Delhi, India
| |
Collapse
|
4
|
Oladipo KH, Parish JL. De-regulation of aurora kinases by oncogenic HPV; implications in cancer development and treatment. Tumour Virus Res 2025; 19:200314. [PMID: 39923999 DOI: 10.1016/j.tvr.2025.200314] [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: 11/29/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025] Open
Abstract
Human papillomaviruses (HPVs) cause diseases ranging from benign warts to invasive cancers. HPVs are the cause of almost all cervical cancers and a sub-set of other epithelial malignancies including head and neck cancers, specifically within the oropharynx. The oncogenic properties of HPV are largely mediated through the viral oncoproteins E6 and E7, which disrupt many cellular pathways to drive uncontrolled cell proliferation. One family of proteins targeted by HPV is the Aurora kinase family. Aurora kinases are serine/threonine kinases including Aurora kinase A (AURKA), B (AURKB), and C (AURKC) which are often dysregulated in many cancer types, including HPV driven cancers. All three family members play essential roles in mitotic regulation and accurate cell division. The deregulation of Aurora kinases by HPV infection highlights their potential as therapeutic targets in HPV-associated malignancies. Targeting Aurora kinase activity, in combination with current HPV therapies, may provide new avenues for treating HPV-induced cancers and reducing the burden of HPV-related diseases. Combinatorial inhibition targets distinct but overlapping functions of these kinases, thereby reducing the potential for cancer cells to develop resistance. This broad impact emphasizes the capability for Aurora kinase inhibitors not only as anti-mitotic agents but also as modulators of multiple oncogenic pathways. This review explores the combinatorial effects of Aurora kinase inhibition, offering insights into novel therapeutic strategies for the treatment of HPV-driven cancers.
Collapse
Affiliation(s)
- Kemi Hannah Oladipo
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom; National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, Birmingham, United Kingdom.
| | - Joanna L Parish
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom; National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, Birmingham, United Kingdom.
| |
Collapse
|
5
|
Tomas EJ, Valdes YR, Davis J, Kolendowski B, Buensuceso A, DiMattia GE, Shepherd TG. Exploiting Cancer Dormancy Signaling Mechanisms in Epithelial Ovarian Cancer Through Spheroid and Organoid Analysis. Cells 2025; 14:133. [PMID: 39851561 PMCID: PMC11764263 DOI: 10.3390/cells14020133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Epithelial ovarian cancer (EOC) exhibits a unique mode of metastasis, involving spheroid formation in the peritoneum. Our research on EOC spheroid cell biology has provided valuable insights into the signaling plasticity associated with metastasis. We speculate that EOC cells modify their biology between tumour and spheroid states during cancer dormancy, although the specific mechanisms underlying this transition remain unknown. Here, we present novel findings from direct comparisons between cultured EOC spheroids and organoids. Our results indicated that AMP-activated protein kinase (AMPK) activity was significantly upregulated and protein kinase B (Akt) was downregulated in EOC spheroids compared to organoids, suggesting a clear differential phenotype. Through RNA sequencing analysis, we further supported these phenotypic differences and highlighted the significance of cell cycle regulation in organoids. By inhibiting the G2/M checkpoint via kinase inhibitors, we confirmed that this pathway is essential for organoids. Interestingly, our results suggest that specifically targeting aurora kinase A (AURKA) may represent a promising therapeutic strategy since our cells were equally sensitive to Alisertib treatment as both spheroids and organoids. Our findings emphasize the importance of studying cellular adaptations of EOC cells, as there may be different therapeutic targets depending on the step of EOC disease progression.
Collapse
Affiliation(s)
- Emily J. Tomas
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
- Department of Anatomy & Cell Biology, Western University, London, ON N6A 5C1, Canada
| | - Yudith Ramos Valdes
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
| | - Jennifer Davis
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
| | - Bart Kolendowski
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
| | - Adrian Buensuceso
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
| | - Gabriel E. DiMattia
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
- Department of Oncology, Western University, London, ON N6A 5C1, Canada
- Department of Biochemistry, Western University, London, ON N6A 5C1, Canada
| | - Trevor G. Shepherd
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
- Department of Anatomy & Cell Biology, Western University, London, ON N6A 5C1, Canada
- Department of Oncology, Western University, London, ON N6A 5C1, Canada
- Department of Obstetrics & Gynaecology, Western University, London, ON N6A 5C1, Canada
| |
Collapse
|
6
|
Zare R, Izadi L, Alarcón-Sánchez MA, Taghva M, Ranjbar MA. Aurora kinase A expression in pleomorphic adenoma, adenoid cystic carcinoma, and mucoepidermoid carcinoma of salivary glands: an immunohistochemical study. BMC Oral Health 2025; 25:89. [PMID: 39825351 PMCID: PMC11740330 DOI: 10.1186/s12903-024-05276-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 11/28/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Aurora kinase A (AurkA) plays a vital role in mitosis and is therefore critical in tumors development and progression. There are a few studies on AurkA expression in salivary gland tumors. The aim of the present study was to evaluate the expression pattern of AurkA in the most common benign and malignant salivary gland tumors by immunohistochemistry. METHODS In this retrospective cross-sectional study, 68 cases including 25 pleomorphic adenomas (PAs), 21 adenoid cystic carcinomas (ADCa), 15 mucoepidermoid carcinomas (MEC), and 7 normal salivary glands (NSG) were enrolled from the archive of the Department of Pathology of Shiraz School of Dentistry, Iran. The expression of AurkA in the tissue samples was assessed by immunohistochemical method and was analyzed using statistical analysis (p < 0.05). RESULTS Of total cases analyzed, the majority of benign and malignant tumors were found to involve minor salivary glands compared to major salivary glands (p < 0.001). In addition, all lesions studied expressed AurkA. More than half of the tumor tissues showed AurkA staining percentages between 26 and 50% and 76-100% compared to NSG (p = 0.08). In 44.1% of cases, cells had a weak staining score, 27.9% a moderate score and the rest (27.9%) a strong score (p = 0.64). CONCLUSION Although AurkA was observed to be expressed in all tumor tissues, further studies are needed to clearly understand the role of AurkA and the possibility of using it as a diagnostic, prognostic and therapeutic factor.
Collapse
Affiliation(s)
- Razieh Zare
- Department of Maxillofacial Pathology, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Izadi
- Undergraduate Student, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mario Alberto Alarcón-Sánchez
- Biomedical Science, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo de los Bravo 39087, Guerrero, Mexico
| | - Masumeh Taghva
- Department of Prosthodontics, School of Dentistry, Shiraz University of Medical Sciences, Qasr-Dasht st., Mehr Intersection, 71956-15878, Shiraz, Iran.
| | - Mohammad Ali Ranjbar
- Department of Maxillofacial Pathology, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Soliman MA, Ahmed HEA, Eltamany EH, Boraei ATA, Aljuhani A, Salama SA, Alghamdi R, Aljohani AKB, Almaghrabi M, Aouad MR. Novel bis-benzimidazole-triazole hybrids: anticancer study, in silico approaches, and mechanistic investigation. Future Med Chem 2025; 17:93-107. [PMID: 39670306 DOI: 10.1080/17568919.2024.2437980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024] Open
Abstract
AIM Benzimidazole-triazole conjugates are very active hotspot for design and synthesis of promising anticancer agents. The target analogs showed potent and selective cytotoxicity over different cancer cell lines for breast and lung ones. MATERIALS & METHODS A new series of bis-1,4-disubstituted-1,2,3-triazoles moieties conjugated with a 2-mercapto-benzimidazole 4a-h and 7a-g was synthesized via the click cycloaddition (CuAAC) reaction. The synthesized triazoles were characterized using several spectroscopic tools. In addition, they were tested against variable cell lines representing different cancer types; HepG-2, MCF-7, HCT-116, and A-549. Computational experiments were introduced for understanding their structure-activity relationships. RESULTS & CONCLUSION The data revealed the outperformance of 7a-g analogs over 4a-h one with very effective IC50 values; 4-13 µg/mL compared to the reference drugs. Moreover, detailed mechanistic analyses showed potent Aurora-A Kinase expression for the most active analogs 7a and 7d exhibiting IC50; 3.5 and 5.3 over the control cells 8 ng/mL respectively. Additionally, based on their Aurora-A Kinase inhibitory activity, compound 7a was promising in apoptosis induction and cell cycle arrest. Molecular docking studies with Aurora-A Kinase revealed binding behaviors similar to the co-crystallized ligand sunitinib. Finally, this scaffold exhibits cytotoxic activity via apoptosis, enzyme downregulation, and suppression of cell division.
Collapse
Affiliation(s)
- Moataz A Soliman
- Deanship of Preparatory Year, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Hany E A Ahmed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Egypt
| | - Elsayed H Eltamany
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Ahmed T A Boraei
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Ateyatallah Aljuhani
- Department of Chemistry, College of Science, Taibah University, Al-Madinah, Al-Munawarah, Saudi Arabia
| | - Samir A Salama
- Division of Biochemistry, Department of Pharmacology, College of Pharmacy, Taif University, Taif, Kingdom of Saudi Arabia
| | - Read Alghamdi
- Pharmacognosy and Pharmaceutical Chemistry Department, Pharmacy College, Taibah University, Al-Madinah, Al-Munawarah, Saudi Arabia
| | - Ahmed K B Aljohani
- Pharmacognosy and Pharmaceutical Chemistry Department, Pharmacy College, Taibah University, Al-Madinah, Al-Munawarah, Saudi Arabia
| | - Mohammed Almaghrabi
- Pharmacognosy and Pharmaceutical Chemistry Department, Pharmacy College, Taibah University, Al-Madinah, Al-Munawarah, Saudi Arabia
| | - Mohamed R Aouad
- Department of Chemistry, College of Science, Taibah University, Al-Madinah, Al-Munawarah, Saudi Arabia
| |
Collapse
|
8
|
Sachdeva A, Roy A, Mandal S. Protein kinase D2-Aurora kinase A-ERK1/2 signalling axis drives neuroendocrine differentiation of epithelial ovarian cancer. Mol Cell Biochem 2025; 480:535-547. [PMID: 38557789 DOI: 10.1007/s11010-024-04986-2] [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: 01/12/2024] [Accepted: 03/09/2024] [Indexed: 04/04/2024]
Abstract
Epithelial ovarian cancer (EOC) is deadliest gynecological malignancy with poor prognosis and patient survival. Despite development of several therapeutic interventions such as poly-ADP ribose polymerase (PARP) inhibitors, EOC remains unmanageable and discovery of novel early detection biomarkers and treatment targets are highly warranted. Although neuroendocrine differentiation (NED) is implicated in different human cancers including prostate adenocarcinoma and lung cancer, mechanistic studies concerning NED of epithelial ovarian cancer are lacking. We report that Aurora kinase A drives NED of epithelial ovarian cancer in an ERK1/2-dependent manner and pharmacological and genetic inhibition of Aurora kinase A suppress NED of ovarian cancer. Moreover, we demonstrate that protein kinase D2 positively regulated Aurora kinase A to drive NED. Overexpression of catalytically active PKD2 drives NED and collectively, PKD2 cross talks with Aurora kinase A/ERK1/2 signalling axis to positively regulate NED of EOC. PKD2/Aurora kinase A/ERK1/2 signalling axis is a novel therapeutic target against neuroendocrine differentiated EOC.
Collapse
Affiliation(s)
- Abha Sachdeva
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Adhiraj Roy
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India.
| | - Supratim Mandal
- Department of Microbiology, University of Kalyani, Kalyani, Nadia, West Bengal, 741235, India
| |
Collapse
|
9
|
Moraes B, Gomes H, Saramago L, Braz V, Parizi LF, Braz G, da Silva Vaz I, Logullo C, Moraes J. Aurora kinase as a putative target to tick control. Parasitology 2024; 151:983-991. [PMID: 39542861 PMCID: PMC11770520 DOI: 10.1017/s003118202400101x] [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: 06/11/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 11/17/2024]
Abstract
Aurora kinases (AURK) play a central role in controlling cell cycle in a wide range of organisms. They belong to the family of serine-threonine kinase proteins. Their role in the cell cycle includes, among others, the entry into mitosis, maturation of the centrosome and formation of the mitotic spindle. In mammals, 3 isoforms have been described: A, B and C, which are distinguished mainly by their function throughout the cell cycle. Two aurora kinase coding sequences have been identified in the transcriptome of the cattle tick Rhipicephalus microplus (Rm-AURKA and Rm-AURKB) containing the aurora kinase-specific domain. For both isoforms, the highest number of AURK coding transcripts is found in ovaries. Based on deduced amino acid sequences, it was possible to identify non-conserved threonine residues which are essential to AURK functions in vertebrates and which are not present in R. microplus sequences. A pan AURK inhibitor (CCT137690) caused cell viability decline in the BME26 tick embryonic cell line. In silico docking assay showed an interaction between Aurora kinase and CCT137690 with exclusive interaction sites in Rm-AURKA. The characterization of exclusive regions of the enzyme will enable new studies aimed at promoting species-specific enzymatic inhibition in ectoparasites.
Collapse
Affiliation(s)
- Bruno Moraes
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil
- Laboratório Integrado de Bioquímica Hatisaburo Masuda, NUPEM-Universidade Federal do Rio de Janeiro campus Macaé, Brazil
| | - Helga Gomes
- Laboratório de Tecido Conjuntivo, Hospital Universitário Clementino Fraga Filho and Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Luiz Saramago
- Laboratório Integrado de Bioquímica Hatisaburo Masuda, NUPEM-Universidade Federal do Rio de Janeiro campus Macaé, Brazil
| | - Valdir Braz
- Laboratório Integrado de Bioquímica Hatisaburo Masuda, NUPEM-Universidade Federal do Rio de Janeiro campus Macaé, Brazil
| | - Luís Fernando Parizi
- Centro de Biotecnologia and Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gloria Braz
- Instituto de Química, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia and Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Carlos Logullo
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Jorge Moraes
- Laboratório Integrado de Bioquímica Hatisaburo Masuda, NUPEM-Universidade Federal do Rio de Janeiro campus Macaé, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
10
|
Sachdeva A, Roy A, Gupta MK, Mandal S. Pharmacological inhibition of protein kinase D2/Aurora kinase A signalling axis suppresses G2/M cell cycle progression and proliferation of epithelial ovarian cancer cells. Pathol Res Pract 2024; 260:155390. [PMID: 38878668 DOI: 10.1016/j.prp.2024.155390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 08/09/2024]
Abstract
Epithelial ovarian cancer (EOC) is the deadliest gynecological malignancy with poor prognosis and patient survival outcome. Protein kinase D2 (PKD2) belongs to Ca++/calmodulin-dependent serine/threonine kinase family and its aberrant expression is associated with many cellular and physiological functions associated with tumorigenesis including cell proliferation. We show that PKD2 is activated during G2/M cell cycle transition and its catalytic inactivation by small molecule inhibitor CRT0066101 or genetic knockdown caused suppression of EOC cell proliferation followed by a delay into mitotic entry. Our RNASeq analysis of PKD2-inactivated EOC cells revealed significant downregulation of genes associated with cell cycle including Aurora kinase A, a critical mitotic regulator. Mechanistically, PKD2 positively regulated Aurora kinase A stability at both transcriptional and post-translational levels by interfering with the function of Fbxw7, drove G2/M cell cycle transition and EOC cell proliferation. Moreover, pharmacological inhibition of Aurora kinase A by small molecule CD532 or its shRNA-mediated genetic knockdown suppressed EOC cell proliferation, induced G2/M cell cycle arrest and mitotic catastrophe followed by apoptosis. Taken together, our results indicated that PKD2 positively regulates Aurora kinase A during G2/M cell cycle entry and pharmacological targeting of PKD2/Aurora kinase A signalling axis could serve as a novel therapeutic intervention against a lethal pathology like EOC.
Collapse
Affiliation(s)
- Abha Sachdeva
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh 201303, India
| | - Adhiraj Roy
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh 201303, India.
| | - Manoj Kumar Gupta
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh 201303, India
| | - Supratim Mandal
- Department of Microbiology, University of Kalyani, Kalyani, Nadia, West Bengal 741235, India
| |
Collapse
|
11
|
Gupta D, Kumar M, Saifi S, Rawat S, Ethayathulla AS, Kaur P. A comprehensive review on role of Aurora kinase inhibitors (AKIs) in cancer therapeutics. Int J Biol Macromol 2024; 265:130913. [PMID: 38508544 DOI: 10.1016/j.ijbiomac.2024.130913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Aurora kinases (AURKs) are a family of serine /threonine protein kinases that have a crucial role in cell cycle process mainly in the event of chromosomal segregation, centrosome maturation and cytokinesis. The family consists of three members including Aurora kinase A (AURK-A), Aurora kinase B (AURK-B) and Aurora kinase C (AURK-C). All AURKs contain a conserved kinase domain for their activity but differ in their cellular localization and functions. AURK-A and AURK-B are expressed mainly in somatic cells while the expression of AURK-C is limited to germ cells. AURK-A promotes G2 to M transition of cell cycle by controlling centrosome maturation and mitotic spindle assembly. AURK-B and AURK-C form the chromosome passenger complex (CPC) that ensures proper chromosomal alignments and segregation. Aberrant expression of AURK-A and AURK-B has been detected in several solid tumours and malignancies. Hence, they have become an attractive therapeutic target against cancer. The first part of this review focuses on AURKs structure, functions, subcellular localization, and their role in tumorigenesis. The review also highlights the functional and clinical impact of selective as well as pan kinase inhibitors. Currently, >60 compounds that target AURKs are in preclinical and clinical studies. The drawbacks of existing inhibitors like selectivity, drug resistance and toxicity have also been addressed. Since, majority of inhibitors are Aurora kinase inhibitor (AKI) type-1 that bind to the active (DFGin and Cin) conformation of the kinase, this information may be utilized to design highly selective kinase inhibitors that can be combined with other therapeutic agents for better clinical outcomes.
Collapse
Affiliation(s)
- Deepali Gupta
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Mukesh Kumar
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Sana Saifi
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Shivani Rawat
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - A S Ethayathulla
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India.
| |
Collapse
|
12
|
Pellecchia S, Viscido G, Franchini M, Gambardella G. Predicting drug response from single-cell expression profiles of tumours. BMC Med 2023; 21:476. [PMID: 38041118 PMCID: PMC10693176 DOI: 10.1186/s12916-023-03182-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/20/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Intra-tumour heterogeneity (ITH) presents a significant obstacle in formulating effective treatment strategies in clinical practice. Single-cell RNA sequencing (scRNA-seq) has evolved as a powerful instrument for probing ITH at the transcriptional level, offering an unparalleled opportunity for therapeutic intervention. RESULTS Drug response prediction at the single-cell level is an emerging field of research that aims to improve the efficacy and precision of cancer treatments. Here, we introduce DREEP (Drug Response Estimation from single-cell Expression Profiles), a computational method that leverages publicly available pharmacogenomic screens from GDSC2, CTRP2, and PRISM and functional enrichment analysis to predict single-cell drug sensitivity from transcriptomic data. We validated DREEP extensively in vitro using several independent single-cell datasets with over 200 cancer cell lines and showed its accuracy and robustness. Additionally, we also applied DREEP to molecularly barcoded breast cancer cells and identified drugs that can selectively target specific cell populations. CONCLUSIONS DREEP provides an in silico framework to prioritize drugs from single-cell transcriptional profiles of tumours and thus helps in designing personalized treatment strategies and accelerating drug repurposing studies. DREEP is available at https://github.com/gambalab/DREEP .
Collapse
Affiliation(s)
- Simona Pellecchia
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Genomics and Experimental Medicine Program, Scuola Superiore Meridionale, Naples, Italy
| | - Gaetano Viscido
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Chemical, Materials and Industrial Engineering, University of Naples Federico II, Naples, Italy
| | - Melania Franchini
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
13
|
Callari M, Sola M, Magrin C, Rinaldi A, Bolis M, Paganetti P, Colnaghi L, Papin S. Cancer-specific association between Tau (MAPT) and cellular pathways, clinical outcome, and drug response. Sci Data 2023; 10:637. [PMID: 37730697 PMCID: PMC10511431 DOI: 10.1038/s41597-023-02543-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/05/2023] [Indexed: 09/22/2023] Open
Abstract
Tau (MAPT) is a microtubule-associated protein causing common neurodegenerative diseases or rare inherited frontotemporal lobar degenerations. Emerging evidence for non-canonical functions of Tau in DNA repair and P53 regulation suggests its involvement in cancer. To bring new evidence for a relevant role of Tau in cancer, we carried out an in-silico pan-cancer analysis of MAPT transcriptomic profile in over 10000 clinical samples from 32 cancer types and over 1300 pre-clinical samples from 28 cancer types provided by the TCGA and the DEPMAP datasets respectively. MAPT expression associated with key cancer hallmarks including inflammation, proliferation, and epithelial to mesenchymal transition, showing cancer-specific patterns. In some cancer types, MAPT functional networks were affected by P53 mutational status. We identified new associations of MAPT with clinical outcomes and drug response in a context-specific manner. Overall, our findings indicate that the MAPT gene is a potential major player in multiple types of cancer. Importantly, the impact of Tau on cancer seems to be heavily influenced by the specific cellular environment.
Collapse
Affiliation(s)
| | - Martina Sola
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Claudia Magrin
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Marco Bolis
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
- Computational Oncology Unit, Department of Oncology, IRCCS Istituto di Ricerche Farmacologiche 'Mario Negri', Milano, Italy
- Swiss Institute of Bioinformatics, Bioinformatics Core Unit, Bellinzona, Switzerland
| | - Paolo Paganetti
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland.
| | - Luca Colnaghi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
| | - Stéphanie Papin
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| |
Collapse
|
14
|
Maciel-Cruz EJ, Figuera-Villanueva LE, Garibaldi-Ríos AF, Gómez-Meda BC, Zúñiga-González GM, Pérez AM, Castro-García PB, Ramírez-Patiño R, Gallegos-Arreola MP. AURKA Gene Variants rs1047972, and rs8173 Are Associated With Breast Cancer. J Breast Cancer 2023; 26:378-390. [PMID: 37565930 PMCID: PMC10475707 DOI: 10.4048/jbc.2023.26.e31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/28/2023] [Accepted: 06/06/2023] [Indexed: 08/12/2023] Open
Abstract
PURPOSE Association between variants rs1047972 and rs8173 of the AURKA gene in healthy women and breast cancer (BC) in a Mexican population. METHODS Genomic DNA samples from 409 healthy women and 572 patients with BC were analyzed for variants rs1047972 and rs8173 of the AURKA gene by polymerase chain reaction-restriction fragment length polymorphism. RESULTS TT genotype (odds ratio [OR], 2.5; 95% confidence interval [CI], 1.22-5.11; p = 0.0015) and the T allele (OR, 1.16; 95% CI, 1.23-2.12; p = 0.0007) of the rs1047972 variant were associated as risk susceptibility for BC relative to the control group. Contrarily, the GG genotype (OR, 0.64; 95% CI, 0.43-0.94; p = 0.029) was associated as a protective factor of susceptibility of BC of the variant rs8173 of the AURKA gene. Differences were observed in the patients with BC who were carriers of the CT genotype of the rs1047972 variant with overweight, obesity, estrogen receptor-positive plus obesity, Ki-67 (≥ 20%) plus history familial positive of cancer; and for variant rs8173 the BC patients who were CG carriers and presented chemotherapy gastric toxicity, hormonal receptor positive plus chemotherapy gastric toxicity, and menopause status plus chemotherapy gastric toxicity (p < 0.05). Two common haplotypes were identified in the study groups: CG and TC genotypes, were associated as a protective and risk factor, respectively (p < 0.05). CONCLUSION Variants rs1047972 and rs8173 of the AURKA gene and the TC haplotype were associated as risk susceptibility factors for BC in this population.
Collapse
Affiliation(s)
- Eric Jonathan Maciel-Cruz
- División de Genética, Centro de Investigación Biomédica de Occidente (CIBO), Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, México
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara (UdeG), Guadalajara, México
| | - Luis Eduardo Figuera-Villanueva
- División de Genética, Centro de Investigación Biomédica de Occidente (CIBO), Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, México
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara (UdeG), Guadalajara, México
| | - Asbiel Felipe Garibaldi-Ríos
- División de Genética, Centro de Investigación Biomédica de Occidente (CIBO), Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, México
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara (UdeG), Guadalajara, México
| | - Belinda Claudia Gómez-Meda
- Departamento de Biología Molecular y Genómica, Instituto de Genética Humana "Dr. Enrique Corona Rivera", Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara (UdeG), Guadalajara, México
| | - Guillermo Moisés Zúñiga-González
- División de Medicina Molecular, Centro de Investigación Biomédica de Occidente (CIBO), Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, México
| | - Ana María Pérez
- Laboratorio de Inmunofarmacología, Centro Universitario de Ciencias Exactas e Ingenierías Universidad de Guadalajara (UdeG), Guadalajara, México
| | - Paola B Castro-García
- Laboratorio de Inmunofarmacología, Centro Universitario de Ciencias Exactas e Ingenierías Universidad de Guadalajara (UdeG), Guadalajara, México
| | - Ramiro Ramírez-Patiño
- Departamento de Medicina y Ciencias de la Vida, Centro Universitario la Ciénega, Universidad de Guadalajara (UdeG), Ocotlán, México
| | - Martha Patricia Gallegos-Arreola
- División de Genética, Centro de Investigación Biomédica de Occidente (CIBO), Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, México.
| |
Collapse
|
15
|
Zhao S, Zhang X, Gao F, Chi H, Zhang J, Xia Z, Cheng C, Liu J. Identification of copper metabolism-related subtypes and establishment of the prognostic model in ovarian cancer. Front Endocrinol (Lausanne) 2023; 14:1145797. [PMID: 36950684 PMCID: PMC10025496 DOI: 10.3389/fendo.2023.1145797] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/10/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is one of the most common and most malignant gynecological malignancies in gynecology. On the other hand, dysregulation of copper metabolism (CM) is closely associated with tumourigenesis and progression. Here, we investigated the impact of genes associated with copper metabolism (CMRGs) on the prognosis of OC, discovered various CM clusters, and built a risk model to evaluate patient prognosis, immunological features, and therapy response. METHODS 15 CMRGs affecting the prognosis of OC patients were identified in The Cancer Genome Atlas (TCGA). Consensus Clustering was used to identify two CM clusters. lasso-cox methods were used to establish the copper metabolism-related gene prognostic signature (CMRGPS) based on differentially expressed genes in the two clusters. The GSE63885 cohort was used as an external validation cohort. Expression of CM risk score-associated genes was verified by single-cell sequencing and quantitative real-time PCR (qRT-PCR). Nomograms were used to visually depict the clinical value of CMRGPS. Differences in clinical traits, immune cell infiltration, and tumor mutational load (TMB) between risk groups were also extensively examined. Tumour Immune Dysfunction and Rejection (TIDE) and Immune Phenotype Score (IPS) were used to validate whether CMRGPS could predict response to immunotherapy in OC patients. RESULTS In the TCGA and GSE63885 cohorts, we identified two CM clusters that differed significantly in terms of overall survival (OS) and tumor microenvironment. We then created a CMRGPS containing 11 genes to predict overall survival and confirmed its reliable predictive power for OC patients. The expression of CM risk score-related genes was validated by qRT-PCR. Patients with OC were divided into low-risk (LR) and high-risk (HR) groups based on the median CM risk score, with better survival in the LR group. The 5-year AUC value reached 0.74. Enrichment analysis showed that the LR group was associated with tumor immune-related pathways. The results of TIDE and IPS showed a better response to immunotherapy in the LR group. CONCLUSION Our study, therefore, provides a valuable tool to further guide clinical management and tailor the treatment of patients with OC, offering new insights into individualized treatment.
Collapse
Affiliation(s)
- Songyun Zhao
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Xin Zhang
- Department of Pathology, The Second People's Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Feng Gao
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Chi
- Southwest Medical University, Luzhou, China
| | | | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians University, Munich, Germany
| | - Chao Cheng
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Bhattacharyya N, Khan MM, Bagabir SA, Almalki AH, Shahwan MA, Haque S, Verma AK, Mangangcha IR. Maximal clique centrality and bottleneck genes as novel biomarkers in ovarian cancer. Biotechnol Genet Eng Rev 2023. [DOI: 10.1080/02648725.2023.2174688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Affiliation(s)
| | - Mohd Mabood Khan
- Division of Molecular Genetics & Biochemistry, National Institute of Cancer Prevention & Research (ICMR-NICPR), Noida, India
| | - Sali Abubaker Bagabir
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Atiah H. Almalki
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
- Addiction and Neuroscience Research Unit, College of Pharmacy, Taif University, Taif, Al-Hawiah, Saudi Arabia
| | - Moyad Al Shahwan
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Ajay Kumar Verma
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | | |
Collapse
|
17
|
SIX3 function in cancer: progression and comprehensive analysis. Cancer Gene Ther 2022; 29:1542-1549. [PMID: 35764712 DOI: 10.1038/s41417-022-00488-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/02/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023]
Abstract
The homeobox gene family encodes transcription factors that are essential for cell growth, proliferation, and differentiation, and its dysfunction is linked to tumor initiation and progression. Sine oculis homeobox (SIX) belongs to the homeobox gene family, with SIX3 being a core member. Recent studies indicate that SXI3 functions as a cancer suppressor or promoter, which is mainly dependent on SIX3's influence on the signal pathways that promote or inhibit cancer in cells. The low expression of SIX3 in most malignant tumors was confirmed by detailed studies, which could promote the cell cycle, proliferation, migration, and angiogenesis. The recovery or upregulation of SIX3 expression to suppress cancer is closely related to the direct or indirect inhibition of the Wnt pathway. However, in some malignancies, such as esophageal cancer and gastric cancer, SIX3 is a tumor-promoting factor, and repressing SIX3 improves patients' prognosis. This review introduces the research progress of SIX3 in tumors and gives a comprehensive analysis, intending to explain why SIX3 plays different roles in different cancers and provide new cancer therapy strategies.
Collapse
|
18
|
Bioinformatic analysis of key pathways and genes shared between endometriosis and ovarian cancer. Arch Gynecol Obstet 2021; 305:1329-1342. [DOI: 10.1007/s00404-021-06285-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 10/13/2021] [Indexed: 12/11/2022]
|
19
|
Sun X, Liu Q, Huang J, Diao G, Liang Z. Transcriptome-based stemness indices analysis reveals platinum-based chemo-theraputic response indicators in advanced-stage serous ovarian cancer. Bioengineered 2021; 12:3753-3771. [PMID: 34266348 PMCID: PMC8806806 DOI: 10.1080/21655979.2021.1939514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Serous ovarian cancer (SOC) is a main histological subtype of ovarian cancer, in which cancer stem cells (CSC) are responsible for its chemoresistance. However, the underlying modulation mechanisms of chemoresistance led by cancer stemness are still undefined. We aimed to investigate potential drug-response indicators among stemness-associated biomarkers in advanced SOC samples. The mRNA expression-based stemness index (mRNAsi) of The Cancer Genome Atlas (TCGA) was evaluated and corrected by tumor purity. Weighted gene co-expression network analysis (WGCNA) was utilized to explore the gene modules and key genes involved in stemness characteristics. We found that mRNAsi and corrected mRNAsi scores were both greater in tumors of Grade 3 and 4 than that of Grade 1 and 2. Forty-two key genes were obtained from the most significant mRNAsi-related gene module. Functional annotation revealed that these key genes were mainly involved in the mitotic division. Thirteen potential platinum-response indicators were selected from the genes enriched to platinum-response associated pathways. Among them, we identified 11 genes with prognostic value of progression-free survival (PFS) in advanced SOC patients treated with platinum and 7 prognostic genes in patients treated with a combination of platinum and taxol. The expressions of the 13 key genes were also validated between platinum-resistant and -sensitive SOC samples of advanced stages in two Gene Expression Omnibus (GEO) datasets. The results revealed that CDC20 was a potential platinum-sensitivity indicator in advanced SOC. These findings may provide a new insight for chemotherapies in advanced SOC patients clinically.
Collapse
Affiliation(s)
- Xinwei Sun
- Department of Gynecology and Obstetrics, Southwest Hospital, Army Medical University, Chongqing, China
| | - Qingyu Liu
- Orthopedic Department, The 964th Hospital of Chinese People's Liberation Army Joint Logistics Support Force, Changchun, China
| | - Jie Huang
- Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing, China
| | - Ge Diao
- Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhiqing Liang
- Department of Gynecology and Obstetrics, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|