1
|
Pai R, Sirigiri DR, Malempati R, Vinjamuri S. Computational investigation of naturally occurring anticancer agents in regulating Hedgehog pathway proteins. PLoS One 2024; 19:e0311307. [PMID: 39625914 PMCID: PMC11614240 DOI: 10.1371/journal.pone.0311307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/11/2024] [Indexed: 12/06/2024] Open
Abstract
Embryonic development in humans is controlled by the Hedgehog pathway, which becomes inactive in mature tissues. Except for tissue maintenance and healing, activation of this pathway results in tumorigenesis with only a few exceptions. The drugs currently in use have shown no effectiveness in blocking the key proteins responsible for tumorigenesis. Therefore, it is crucial to find new inhibitors that can stop the abnormal activation of the pathway. A preliminary Insilco screening of naturally occurring compounds was carried out to identify potential inhibitors of the pathway. Docking of seventeen naturally occurring antitumorigenic compounds against the four key proteins of the regulatory proteins of the Hedgehog pathway using AutoDock v4.2.6 software was carried out. Liriodenine exhibited the strongest binding affinity towards three out of the four regulatory proteins (-7.61 kcal/mol with Smoothened, -8.14 kcal/mol with Patched-I, and -6.15 kcal/mol with Gli-II) of the Hedgehog pathway, whereas 2',4-dihydroxy-3-methoxychalcone displayed the highest binding affinity of -7.04 kcal/mol with the Sonic Hedgehog protein. Additional molecular dynamic simulation was conducted using Gromacs with Liriodenine and 2',4-dihydroxy-3-methoxy chalcone. Every protein-ligand complex underwent simulation using v5.1.4 software for a duration of 100 nanoseconds. The findings from the simulation indicate that Liriodenine and 2',4-dihydroxy-3-methoxy chalcone form a strong bond with their corresponding protein. Our findings show that the two aforementioned molecules have potential as new inhibitors of the pathway and should be further investigated in both invitro and in vivo experiments.
Collapse
Affiliation(s)
- Renu Pai
- Department of Biotechnology, BMS College of Engineering, Bengaluru, Karnataka, India
| | | | | | - Saisha Vinjamuri
- Department of Biotechnology, BMS College of Engineering, Bengaluru, Karnataka, India
| |
Collapse
|
2
|
Yang L, Wang X, Lin Q, Shen G, Chen H. CLDN11 deficiency upregulates FOXM1 to facilitate breast tumor progression through hedgehog signaling pathway. J Mol Histol 2024; 55:1259-1270. [PMID: 39438406 PMCID: PMC11567981 DOI: 10.1007/s10735-024-10267-5] [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: 06/28/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024]
Abstract
Claudins (CLDNs) play a crucial role in regulating the permeability of epithelial barriers and can impact tumor behavior through alterations in their expression. However, the precise mechanisms underlying the involvement of CLDNs in breast cancer progression remain unclear. This study aimed to investigate the role of CLDN11 in breast cancer progression. Utilizing the TCGA database and clinical specimens from breast cancer patients, we observed reduced expression of CLDN11 in tumor tissues, which correlated with poor prognosis in breast cancer patients. In vitro, silencing of CLDN11 enhanced the proliferative and migratory characteristics of breast cancer cell lines MCF-7 and MDA-MB-231. Mechanistically, CLDN11 deficiency promoted the upregulation of Forkhead Box M1 (FOXM1) by activating the hedgehog signaling pathway, thereby sustaining tumor progression in breast cancer. In vivo, blockade of hedgehog signaling suppressed the tumor progression induced by CLDN11 silencing. Our study highlights the significance of the CLDN11/FOXM1 axis in breast cancer progression, suggesting CLDN11 as a potential diagnostic indicator and therapeutic target for clinical therapy.
Collapse
Affiliation(s)
- Leyi Yang
- Department of Breast Surgery, Zhangzhou Hospital, Fujian Medical University, No 59 Shengli West Road, Xiangcheng district, Zhangzhou, Fujian province, 363000, China
| | - Xiaoping Wang
- Department of Breast Surgery, Zhangzhou Hospital, Fujian Medical University, No 59 Shengli West Road, Xiangcheng district, Zhangzhou, Fujian province, 363000, China
| | - Qinghai Lin
- Department of Breast Surgery, Zhangzhou Hospital, Fujian Medical University, No 59 Shengli West Road, Xiangcheng district, Zhangzhou, Fujian province, 363000, China.
| | - Guoyi Shen
- Department of Thoracic Surgery, Zhangzhou Hospital, Fujian Medical University, Zhangzhou, China
| | - Hong Chen
- Department of Ultrasound, Zhangzhou Hospital, Fujian Medical University, Zhangzhou, China
| |
Collapse
|
3
|
Sai BM, Dinakar YH, Kumar H, Jain R, Kesharwani S, Kesharwani SS, Mudavath SL, Ramkishan A, Jain V. Therapeutic delivery of siRNA for the management of breast cancer and triple-negative breast cancer. Ther Deliv 2024; 15:871-891. [PMID: 39320858 PMCID: PMC11498026 DOI: 10.1080/20415990.2024.2400044] [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: 05/24/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Breast cancer is the leading cause of cancer-related deaths among women globally. The difficulties with anticancer medications, such as ineffective targeting, larger doses, toxicity to healthy cells and side effects, have prompted attention to alternate approaches to address these difficulties. RNA interference by small interfering RNA (siRNA) is one such tactic. When compared with chemotherapy, siRNA has several advantages, including the ability to quickly modify and suppress the expression of the target gene and display superior efficacy and safety. However, there are known challenges and hurdles that limits their clinical translation. Decomposition by endonucleases, renal clearance, hydrophilicity, negative surface charge, short half-life and off-target effects of naked siRNA are obstacles that hinder the desired biological activity of naked siRNA. Nanoparticulate systems such as polymeric, lipid, lipid-polymeric, metallic, mesoporous silica nanoparticles and several other nanocarriers were used for effective delivery of siRNA and to knock down genes involved in breast cancer and triple-negative breast cancer. The focus of this review is to provide a comprehensive picture of various strategies utilized for delivering siRNA, such as combinatorial delivery, development of modified nanoparticles, smart nanocarriers and nanocarriers that target angiogenesis, cancer stem cells and metastasis of breast cancer.
Collapse
Affiliation(s)
- Boya Manasa Sai
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Yirivinti Hayagreeva Dinakar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Sharyu Kesharwani
- National Institute of Pharmaceutical Education & Research, Kolkata, West Bengal , 700054, India
| | | | - Shyam lal Mudavath
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Ajmeer Ramkishan
- Central Drugs Standard Control Organization, East Zone, Kolkata, 700020, West Bengal, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| |
Collapse
|
4
|
Liu R, Yu Y, Wang Q, Zhao Q, Yao Y, Sun M, Zhuang J, Sun C, Qi Y. Interactions between hedgehog signaling pathway and the complex tumor microenvironment in breast cancer: current knowledge and therapeutic promises. Cell Commun Signal 2024; 22:432. [PMID: 39252010 PMCID: PMC11382420 DOI: 10.1186/s12964-024-01812-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/31/2024] [Indexed: 09/11/2024] Open
Abstract
Breast cancer ranks as one of the most common malignancies among women, with its prognosis and therapeutic efficacy heavily influenced by factors associated with the tumor cell biology, particularly the tumor microenvironment (TME). The diverse elements of the TME are engaged in dynamic bidirectional signaling interactions with various pathways, which together dictate the growth, invasiveness, and metastatic potential of breast cancer. The Hedgehog (Hh) signaling pathway, first identified in Drosophila, has been established as playing a critical role in human development and disease. Notably, the dysregulation of the Hh pathway is recognized as a major driver in the initiation, progression, and metastasis of breast cancer. Consequently, elucidating the mechanisms by which the Hh pathway interacts with the distinct components of the breast cancer TME is essential for comprehensively evaluating the link between Hh pathway activation and breast cancer risk. This understanding is also imperative for devising novel targeted therapeutic strategies and preventive measures against breast cancer. In this review, we delineate the current understanding of the impact of Hh pathway perturbations on the breast cancer TME, including the intricate and complex network of intersecting signaling cascades. Additionally, we focus on the therapeutic promise and clinical challenges of Hh pathway inhibitors that target the TME, providing insights into their potential clinical utility and the obstacles that must be overcome to harness their full therapeutic potential.
Collapse
Affiliation(s)
- Ruijuan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China
| | - Yang Yu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China
| | - Qingyang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Qianxiang Zhao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China
| | - Mengxuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China.
| | - Yuanfu Qi
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
5
|
Parambil ST, Antony GR, Littleflower AB, Subhadradevi L. The molecular crosstalk of the hippo cascade in breast cancer: A potential central susceptibility. Biochimie 2024; 222:132-150. [PMID: 38494109 DOI: 10.1016/j.biochi.2024.03.008] [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: 01/22/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
The incidence of breast cancer is perpetually growing globally, and it remains a major public health problem and the leading cause of mortality in women. Though the aberrant activities of the Hippo pathway have been reported to be associated with cancer, constructive knowledge of the pathway connecting the various elements of breast cancer remains to be elucidated. The Hippo transducers, yes-associated protein (YAP) and transcriptional co-activator with PDZ binding motif (TAZ), are reported to be either tumor suppressors, oncogenes, or independent prognostic markers in breast cancer. Thus, there is further need for an explicative evaluation of the dilemma with this molecular contribution of Hippo transducers in modulating breast malignancy. In this review, we summarize the intricate crosstalk of the Hippo pathway in different aspects of breast malignancy, including stem-likeness, cellular signaling, metabolic adaptations, tumor microenvironment, and immune responses. The collective data shows that Hippo transducers play an indispensable role in mammary tumor formation, progression, and dissemination. However, the cellular functions of YAP/TAZ in tumorigenesis might be largely dependent on the mechanical and biophysical cues they interact with, as well as on the cell phenotype. This review provides a glimpse into the plausible biological contributions of the cascade to the inward progression of breast carcinoma and suggests potential therapeutic prospects.
Collapse
Affiliation(s)
- Sulfath Thottungal Parambil
- Laboratory of Molecular Medicine, Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, 695011, Kerala, India
| | - Gisha Rose Antony
- Laboratory of Molecular Medicine, Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, 695011, Kerala, India
| | - Ajeesh Babu Littleflower
- Laboratory of Molecular Medicine, Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, 695011, Kerala, India
| | - Lakshmi Subhadradevi
- Laboratory of Molecular Medicine, Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, 695011, Kerala, India.
| |
Collapse
|
6
|
Fan Z, Pan H, Qu N, Wang X, Cao L, Chen L, Liu M. LncRNA taurine upregulated gene 1 in liver disease. Clin Chim Acta 2024; 560:119752. [PMID: 38821337 DOI: 10.1016/j.cca.2024.119752] [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: 04/10/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Long non-coding RNAs (lncRNAs) are RNA sequences exceeding 200 nucleotides in length that lack protein-coding capacity and participate in diverse biological processes in the human body, particularly exerting a pivotal role in disease surveillance, diagnosis, and progression. Taurine upregulated gene 1 (TUG1) is a versatile lncRNA, and recent studies have revealed that the aberrant expression or function of TUG1 is intricately linked to the pathogenesis of liver diseases. Consequently, we have summarized the current understanding of the mechanism of TUG1 in liver diseases such as liver fibrosis, fatty liver, cirrhosis, liver injury, hepatitis, and liver cancer. Moreover, mounting evidence suggests that interventions targeting TUG1 or its downstream pathways may hold therapeutic promise for liver diseases. This review elucidates the characteristics, mechanisms, and targets of TUG1 in liver diseases, offering a theoretical basis for the prevention, diagnosis, treatment, and prognostic biomarkers of liver diseases.
Collapse
Affiliation(s)
- Zihao Fan
- School of Pharmaceutical Sciences, Liaoning University, No. 66, Chongshan Mid Road, Shenyang 110036, China
| | - Hao Pan
- School of Pharmaceutical Sciences, Liaoning University, No. 66, Chongshan Mid Road, Shenyang 110036, China
| | - Na Qu
- School of Pharmaceutical Sciences, Liaoning University, No. 66, Chongshan Mid Road, Shenyang 110036, China
| | - Xin Wang
- School of Pharmaceutical Sciences, Liaoning University, No. 66, Chongshan Mid Road, Shenyang 110036, China
| | - Lianrui Cao
- School of Pharmaceutical Sciences, Liaoning University, No. 66, Chongshan Mid Road, Shenyang 110036, China
| | - Lijiang Chen
- School of Pharmaceutical Sciences, Liaoning University, No. 66, Chongshan Mid Road, Shenyang 110036, China.
| | - Mingxia Liu
- School of Pharmaceutical Sciences, Liaoning University, No. 66, Chongshan Mid Road, Shenyang 110036, China.
| |
Collapse
|
7
|
Noser AA, El-Barbary AA, Salem MM, El Salam HAA, Shahien M. Synthesis and molecular docking simulations of novel azepines based on quinazolinone moiety as prospective antimicrobial and antitumor hedgehog signaling inhibitors. Sci Rep 2024; 14:3530. [PMID: 38347004 PMCID: PMC10861550 DOI: 10.1038/s41598-024-53517-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/01/2024] [Indexed: 02/15/2024] Open
Abstract
A series of novel azepine derivatives based on quinazolinone moiety was synthesized through the reaction of quinazolinone chalcones (2a-d) either with 2-amino aniline in acidic medium to give diazepines (3a-d) or with 2-aminophenol to offer oxazepine (4a-d). The structure of the synthesized compounds was confirmed via melting points, elemental analyses, and different spectroscopic techniques. Moreover, these newly compounds mode of action was investigated in-silico using molecular docking against the outer membrane protein A (OMPA), exo-1,3-beta-glucanase for their antimicrobial activity, and against Smoothened (SMO), transcription factor glioma-associated homology (SUFU/GLI-1), the main proteins of Hedgehog signaling pathway to inspect their anticancer potential. Our results showed that, diazepine (3a) and oxazepine (4a) offered the highest binding energy against the target OMPA/ exo-1,3-beta-glucanase proteins and exhibited the potent antimicrobial activities against E. coli, P. aeruginosa, S. aureus, B. subtilis, C. Albicans and A. flavus. As well, diazepine (3a) and oxazepine (4a) achieved the best results among the other compounds, in their binding energy against the target SMO, SUFU/GLI-1 proteins. The in-vitro cytotoxic study was done for them on panel of cancer cell lines HCT-116, HepG2, and MCF-7 and normal cell line WI-38. Conclusively, it was revealed that molecular docking in-silico simulations and the in-vitro experiments were agreed. As a result, our findings elucidated that diazepine (3a) and oxazepine (4a), have the potential to be used as antimicrobial agents and as possible cancer treatment medications.
Collapse
Affiliation(s)
- Ahmed A Noser
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - A A El-Barbary
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Maha M Salem
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Hayam A Abd El Salam
- Green Chemistry Department, National Research Centre, Dokki, GizaCairo, 12622, Egypt
| | - Mohamed Shahien
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
8
|
Srinivasan M, Gangurde A, Chandane AY, Tagalpallewar A, Pawar A, Baheti AM. Integrating network pharmacology and in silico analysis deciphers Withaferin-A's anti-breast cancer potential via hedgehog pathway and target network interplay. Brief Bioinform 2024; 25:bbae032. [PMID: 38446743 PMCID: PMC10917074 DOI: 10.1093/bib/bbae032] [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: 09/07/2023] [Revised: 12/09/2023] [Accepted: 12/22/2023] [Indexed: 03/08/2024] Open
Abstract
This study examines the remarkable effectiveness of Withaferin-A (WA), a withanolide obtained from Withania somnifera (Ashwagandha), in encountering the mortiferous breast malignancy, a global peril. The predominant objective is to investigate WA's intrinsic target proteins and hedgehog (Hh) pathway proteins in breast cancer targeting through the application of in silico computational techniques and network pharmacology predictions. The databases and webtools like Swiss target prediction, GeneCards, DisGeNet and Online Mendelian Inheritance in Man were exploited to identify the common target proteins. The culmination of the WA network and protein-protein interaction network were devised using Stitch and String web tools, through which the drug-target network of 30 common proteins was constructed employing Cytoscape-version 3.9. Enrichment analysis was performed by incorporating Gprofiler, Metascape and Cytoscape plugins. David compounded the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, and enrichment was computed through bioinformatics tools. The 20 pivotal proteins were docked harnessing Glide, Schrodinger Suite 2023-2. The investigation was governed by docking scores and affinity. The shared target proteins underscored the precise Hh and WA network roles with the affirmation enrichment P-value of <0.025. The implications for hedgehog and cancer pathways were profound with enrichment (P < 0.01). Further, the ADMET and drug-likeness assessments assisted the claim. Robust interactions were noticed with docking studies, authenticated through molecular dynamics, molecular mechanics generalized born surface area scores and bonds. The computational investigation emphasized WA's credible anti-breast activity, specifically with Hh proteins, implying stem-cell-level checkpoint restraints. Rigorous testament is imperative through in vitro and in vivo studies.
Collapse
Affiliation(s)
- Mythili Srinivasan
- Research Scholar, School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, Maharashtra 411038, India
| | - Apeksha Gangurde
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, Maharashtra 411038, India
| | - Ashwini Y Chandane
- Abhinav College of Pharmacy, Narhe, Ambegaon, Pune, Maharashtra 411041, India
| | - Amol Tagalpallewar
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, Maharashtra 411038, India
| | - Anil Pawar
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, Maharashtra 411038, India
| | - Akshay M Baheti
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, Maharashtra 411038, India
| |
Collapse
|
9
|
Mohammed OA, Doghish AS, Saleh LA, Alghamdi M, Alamri MMS, Alfaifi J, Adam MIE, Alharthi MH, Alshahrani AM, Alhalafi AH, BinAfif WF, Rezigalla AA, Abdel-Reheim MA, El-Wakeel HS, Attia MA, Elmorsy EA, Al-Noshokaty TM, Nomier Y, Saber S. Itraconazole halts hepatocellular carcinoma progression by modulating sonic hedgehog signaling in rats: A novel therapeutic approach. Pathol Res Pract 2024; 253:155086. [PMID: 38176308 DOI: 10.1016/j.prp.2023.155086] [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: 12/09/2023] [Revised: 12/24/2023] [Accepted: 12/30/2023] [Indexed: 01/06/2024]
Abstract
Liver cancer stands as the fourth leading global cause of death, and its prognosis remains grim due to the limited effectiveness of current medical interventions. Among the various pathways implicated in the development of hepatocellular carcinoma (HCC), the hedgehog signaling pathway has emerged as a crucial player. Itraconazole, a relatively safe and cost-effective antifungal medication, has gained attention for its potential as an anticancer agent. Its primary mode of action involves inhibiting the hedgehog pathway, yet its impact on HCC has not been elucidated. The main objective of this study was to investigate the effect of itraconazole on diethylnitrosamine-induced early-stage HCC in rats. Our findings revealed that itraconazole exhibited a multifaceted arsenal against HCC by downregulating the expression of key components of the hedgehog pathway, shh, smoothened (SMO), and GLI family zinc finger 1 (GLI1), and GLI2. Additionally, itraconazole extended survival and improved liver tissue structure, attributed mainly to its inhibitory effects on hedgehog signaling. Besides, itraconazole demonstrated a regulatory effect on Notch1, and Wnt/β-catenin signaling molecules. Consequently, itraconazole displayed diverse anticancer properties, including anti-inflammatory, antiangiogenic, antiproliferative, and apoptotic effects, as well as the potential to induce autophagy. Moreover, itraconazole exhibited a promise to impede the transformation of epithelial cells into a more mesenchymal-like phenotype. Overall, this study emphasizes the significance of targeting the hedgehog pathway with itraconazole as a promising avenue for further exploration in clinical studies related to HCC treatment.
Collapse
Affiliation(s)
- Osama A Mohammed
- Department of Pharmacology, College of medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo 11231, Egypt.
| | - Lobna A Saleh
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt; Department of Pharmacology and Toxicology, Collage of Pharmacy, Taif University, Taif, Saudi Arabia.
| | - Mushabab Alghamdi
- Department of Internal Medicine, Division of Rheumatology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Mohannad Mohammad S Alamri
- Department of Family and Community Medicine, College of medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Jaber Alfaifi
- Department of Child Health, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Masoud I E Adam
- Department of Medical Education and Internal Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Muffarah Hamid Alharthi
- Department of Family and Community Medicine, College of medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Abdullah M Alshahrani
- Department of Family and Community Medicine, College of medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Abdullah Hassan Alhalafi
- Department of Family and Community Medicine, College of medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Waad Fuad BinAfif
- Department of Internal Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Assad Ali Rezigalla
- Department of Anatomy, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Hend S El-Wakeel
- Physiology Department, Benha Faculty of Medicine, Benha University, Qalubyia 13518, Egypt; Physiology Department, Al-Baha Faculty of Medicine, Al-Baha University, Al-Baha 65799, Saudi Arabia.
| | - Mohammed A Attia
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; Department of Basic Medical Sciences , College of Medicine Almaarefa University Diriyiah, 13713, Riyadh, Saudi Arabia.
| | - Elsayed A Elmorsy
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; Pharmacology and Therapeutics Department, Qassim College of Medicine, Qassim University, Buraydah 51452, Saudi Arabia.
| | - Tohada M Al-Noshokaty
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt.
| | - Yousra Nomier
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman.
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| |
Collapse
|
10
|
Dinakar YH, Rajana N, Kumari NU, Jain V, Mehra NK. Recent Advances of Multifunctional PLGA Nanocarriers in the Management of Triple-Negative Breast Cancer. AAPS PharmSciTech 2023; 24:258. [PMID: 38097825 DOI: 10.1208/s12249-023-02712-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Even though chemotherapy stands as a standard option in the therapy of TNBC, problems associated with it such as anemia, bone marrow suppression, immune suppression, toxic effects on healthy cells, and multi-drug resistance (MDR) can compromise their effects. Nanoparticles gained paramount importance in overcoming the limitations of conventional chemotherapy. Among the various options, nanotechnology has appeared as a promising path in preclinical and clinical studies for early diagnosis of primary tumors and metastases and destroying tumor cells. PLGA has been extensively studied amongst various materials used for the preparation of nanocarriers for anticancer drug delivery and adjuvant therapy because of their capability of higher encapsulation, easy surface functionalization, increased stability, protection of drugs from degradation versatility, biocompatibility, and biodegradability. Furthermore, this review also provides an overview of PLGA-based nanoparticles including hybrid nanoparticles such as the inorganic PLGA nanoparticles, lipid-coated PLGA nanoparticles, cell membrane-coated PLGA nanoparticles, hydrogels, exosomes, and nanofibers. The effects of all these systems in various in vitro and in vivo models of TNBC were explained thus pointing PLGA-based NPs as a strategy for the management of TNBC.
Collapse
Affiliation(s)
- Yirivinti Hayagreeva Dinakar
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India
| | - Naveen Rajana
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India
| | - Nalla Usha Kumari
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India.
| |
Collapse
|
11
|
Guo Z, Han S. Targeting cancer stem cell plasticity in triple-negative breast cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:1165-1181. [PMID: 38213533 PMCID: PMC10776602 DOI: 10.37349/etat.2023.00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/15/2023] [Indexed: 01/13/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype with limited treatment options. Cancer stem cells (CSCs) are thought to play a crucial role in TNBC progression and resistance to therapy. CSCs are a small subpopulation of cells within tumors that possess self-renewal and differentiation capabilities and are responsible for tumor initiation, maintenance, and metastasis. CSCs exhibit plasticity, allowing them to switch between states and adapt to changing microenvironments. Targeting CSC plasticity has emerged as a promising strategy for TNBC treatment. This review summarizes recent advances in understanding the molecular mechanisms underlying CSC plasticity in TNBC and discusses potential therapeutic approaches targeting CSC plasticity.
Collapse
Affiliation(s)
- Zhengwang Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shuyan Han
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
12
|
Yin L, Cao R, Liu Z, Luo G, Li Y, Zhou X, Chen X, Wu Y, He J, Zu X, Shen Y. FUNDC2, a mitochondrial outer membrane protein, mediates triple-negative breast cancer progression via the AKT/GSK3β/GLI1 pathway. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1770-1783. [PMID: 37700593 PMCID: PMC10679879 DOI: 10.3724/abbs.2023142] [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: 04/06/2023] [Accepted: 05/12/2023] [Indexed: 09/14/2023] Open
Abstract
Triple-negative breast cancer (TNBC) lacks effective therapeutic targets and has a poor prognosis, easy recurrence and metastasis. It is urgent and important to explore TNBC treatment targets. Through mass spectrometry combined with qRT-PCR validation in luminal A cells and TNBC cells, high-content screening and clinical sample analysis, FUNDC2 was discovered as a novel target. The function of the outer mitochondrial membrane protein FUNDC2 in breast cancer is still unclear. In this study, we find that FUNDC2 expression in TNBC tissues is significantly higher than that in luminal subtype breast cancer tissues. FUNDC2 silencing in TNBC cells significantly reduces cell proliferation, migration and invasion. As demonstrated in vivo using subcutaneous tumor xenografts in mice, FUNDC2 suppression significantly inhibits tumor growth. The underlying mechanism might be mediated by inactivating its downstream signal AKT/GSK3β and GLI1, a key factor of the Hedgehog signaling pathway. Therefore, FUNDC2 may promote TNBC progression and provide a therapeutic target for treating TNBC.
Collapse
Affiliation(s)
- Liyang Yin
- The First Affiliated HospitalCancer Research InstituteHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Renxian Cao
- The First Affiliated HospitalCancer Research InstituteHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Zhuoqing Liu
- The First Affiliated HospitalCancer Research InstituteHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Gang Luo
- The First Affiliated HospitalCancer Research InstituteHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Yu Li
- The First Affiliated HospitalCancer Research InstituteHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Xiaolong Zhou
- The First Affiliated HospitalCancer Research InstituteHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Xiguang Chen
- The First Affiliated HospitalCancer Research InstituteHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Ying Wu
- The First Affiliated HospitalCancer Research InstituteHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Jun He
- Nanhua Affiliated HospitalDepartment of Spine SurgeryHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Xuyu Zu
- The First Affiliated HospitalCancer Research InstituteHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Yingying Shen
- The First Affiliated HospitalCancer Research InstituteHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| |
Collapse
|
13
|
Wang X, Xu J, Sun Y, Cao S, Zeng H, Jin N, Shou M, Tang S, Chen Y, Huang M. Hedgehog pathway orchestrates the interplay of histone modifications and tailors combination epigenetic therapies in breast cancer. Acta Pharm Sin B 2023; 13:2601-2612. [PMID: 37425067 PMCID: PMC10326305 DOI: 10.1016/j.apsb.2023.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/09/2023] [Accepted: 02/28/2023] [Indexed: 07/11/2023] Open
Abstract
Epigenetic therapies that cause genome-wide epigenetic alterations, could trigger local interplay between different histone marks, leading to a switch of transcriptional outcome and therapeutic responses of epigenetic treatment. However, in human cancers with diverse oncogenic activation, how oncogenic pathways cooperate with epigenetic modifiers to regulate the histone mark interplay is poorly understood. We herein discover that the hedgehog (Hh) pathway reprograms the histone methylation landscape in breast cancer, especially in triple-negative breast cancer (TNBC). This facilitates the histone acetylation caused by histone deacetylase (HDAC) inhibitors and gives rise to new therapeutic vulnerability of combination therapies. Specifically, overexpression of zinc finger protein of the cerebellum 1 (ZIC1) in breast cancer promotes Hh activation, facilitating the switch of H3K27 methylation (H3K27me) to acetylation (H3K27ac). The mutually exclusive relationship of H3K27me and H3K27ac allows their functional interplay at oncogenic gene locus and switches therapeutic outcomes. Using multiple in vivo breast cancer models including patient-derived TNBC xenograft, we show that Hh signaling-orchestrated H3K27me and H3K27ac interplay tailors combination epigenetic drugs in treating breast cancer. Together, this study reveals the new role of Hh signaling-regulated histone modifications interplay in responding to HDAC inhibitors and suggests new epigenetically-targeted therapeutic solutions for treating TNBC.
Collapse
Affiliation(s)
- Xiaomin Wang
- Cancer Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Xu
- Cancer Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiming Sun
- Cancer Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siyuwei Cao
- Cancer Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hanlin Zeng
- Cancer Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Nan Jin
- Cancer Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Matthew Shou
- Division of Diabetes, Endocrinology and Metabolism, Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Shuai Tang
- Cancer Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yi Chen
- Cancer Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Min Huang
- Cancer Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
14
|
1'-O-methyl-averantin isolated from the endolichenic fungus Jackrogersella sp. EL001672 suppresses colorectal cancer stemness via sonic Hedgehog and Notch signaling. Sci Rep 2023; 13:2811. [PMID: 36797277 PMCID: PMC9935543 DOI: 10.1038/s41598-023-28773-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/24/2023] [Indexed: 02/18/2023] Open
Abstract
Endolichenic fungi are host organisms that live on lichens and produce a wide variety of secondary metabolites. Colorectal cancer stem cells are capable of self-renewal and differentiation into cancer cells, which makes cancers difficult to eradicate. New alternative therapeutics are needed to inhibit the growth of tumor stem cells. This study examined the ability of an extract of Jackrogersella sp. EL001672 (derived from the lichen Cetraria sp.) and the isolated compound 1'-O-methyl-averantin to inhibit development of cancer stemness. The endolichenic fungus Jackrogersella sp. EL001672 (KACC 83021BP), derived from Cetraria sp., was grown in culture medium. The culture broth was extracted with acetone to obtain a crude extract. Column chromatography and reverse-phase HPLC were used to isolate an active compound. The anticancer activity of the extract and the isolated compound was evaluated by qRT-PCR and western blotting, and in cell viability, spheroid formation, and reporter assays. The acetone extract of EL001672 did not affect cell viability. However, 1'-O-methyl-averantin showed cytotoxic effects against cancer cell lines at 50 μg/mL and 25 μg/mL. Both the crude extract and 1'-O-methyl-averantin suppressed spheroid formation in CRC cell lines, and downregulated expression of stemness markers ALDH1, CD44, CD133, Lgr-5, Msi-1, and EphB1. To further characterize the mechanism underlying anti-stemness activity, we examined sonic Hedgehog and Notch signaling. The results showed that the crude extract and the 1'-O-methyl-averantin inhibited Gli1, Gli2, SMO, Bmi-1, Notch-1, Hes-1, and the CSL complex. Consequently, an acetone extract and 1'-O-methyl-averantin isolated from EL001672 suppresses colorectal cancer stemness by regulating the sonic Hedgehog and Notch signaling pathways.
Collapse
|
15
|
FBXL2 promotes E47 protein instability to inhibit breast cancer stemness and paclitaxel resistance. Oncogene 2023; 42:339-350. [PMID: 36460773 DOI: 10.1038/s41388-022-02559-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with a high risk of metastasis and recurrence. Although chemotherapy has greatly improved the clinical outcome of TNBC patients, acquired drug resistance remains a huge challenge for TNBC treatment. Breast cancer stem cells (BCSCs) play a critical role in breast cancer development, metastasis, recurrence, and chemotherapy resistance. Thus, it is of great importance to decipher the underlying molecular mechanism of BCSCs regulation for TNBC drug resistance. In this study, we demonstrate that the F-box protein FBXL2 is a critical negative regulator of BCSCs stemness and that downregulation of FBXL2 plays a causal role in TNBC drug resistance. We show that expression levels of FBXL2 significantly influence CD44high/CD24low subpopulation and the mammosphere formation ability of TNBC cells. Ectopic expression of FBXL2 inhibits initiation of TNBC and overcomes paclitaxel resistance in vivo. In addition, activation of FBXL2 by nebivolol, a clinically used small-molecule inhibitor of the beta-1 receptor, markedly overcomes BCSCs-induced paclitaxel resistance. Mechanistically, we show that FBXL2 targets transcriptional factor E47 for polyubiquitin- and proteasome-mediated degradation, resulting in inhibition of BCSC stemness. Clinical analyses indicate that low expression of FBXL2 correlates with high expression of E47 as well as with high stemness features, and is associated with poor clinical outcomes of breast cancer patients. Taken together, these results highlight that the FBXL2-E47 axis plays a critical role in the regulation of BCSC stemness and paclitaxel resistance. Thus, targeting FBXL2 might be a potential therapeutic strategy for drug-resistant TNBC.
Collapse
|
16
|
Furukawa N, Stearns V, Santa-Maria CA, Popel AS. The tumor microenvironment and triple-negative breast cancer aggressiveness: shedding light on mechanisms and targeting. Expert Opin Ther Targets 2022; 26:1041-1056. [PMID: 36657483 PMCID: PMC10189896 DOI: 10.1080/14728222.2022.2170779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
INTRODUCTION In contrast to other breast cancer subtypes, there are currently limited options of targeted therapies for triple-negative breast cancer (TNBC). Immense research has demonstrated that not only cancer cells but also stromal cells and immune cells in the tumor microenvironment (TME) play significant roles in the progression of TNBC. It is thus critical to understand the components of the TME of TNBC and the interactions between the various cell populations. AREAS COVERED The components of the TME of TNBC identified by single-cell technologies are reviewed. Furthermore, the molecular interactions between the cells and the potential therapeutic targets contributing to the progression of TNBC are discussed. EXPERT OPINION Single-cell omics studies have contributed to the classification of cells in the TME and the identification of important cell types involved in the progression and the treatment of the tumor. The interactions between cancer cells and stromal cells/immune cells in the TME have led to the discovery of potential therapeutic targets. Experimental data with spatial and temporal resolution will further boost the understanding of the TME of TNBC.
Collapse
Affiliation(s)
- Natsuki Furukawa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Vered Stearns
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Cesar A. Santa-Maria
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
17
|
Budimir I, Tomasović-Lončarić Č, Kralik K, Čonkaš J, Eljuga D, Žic R, Gorjanc B, Tucaković H, Caktaš D, Jaman J, Lisek V, Vlajčić Z, Martić K, Ozretić P. Higher Expressions of SHH and AR Are Associated with a Positive Receptor Status and Have Impact on Survival in a Cohort of Croatian Breast Cancer Patients. Life (Basel) 2022; 12:life12101559. [PMID: 36294994 PMCID: PMC9605052 DOI: 10.3390/life12101559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancers (BC) are usually classified into four molecular subtypes according to the expression of estrogen (ER), progesterone (PR), and human epidermal growth factor 2 (HER2) receptors and proliferation marker Ki-67. Despite available anti-hormonal therapies and due to the inherent propensity of some subtypes to develop metastasis, there is a permanent need to discover new prognostic and predictive biomarkers, as well as therapeutic targets for BC. In this study, we used immunohistochemical staining to determine the expression of androgen receptor (AR) and sonic hedgehog protein (SHH), the main ligand of the Hedgehog-GLI (HH-GLI) signaling pathway, in 185 archival primary BC tissue samples and correlated it with clinicopathological characteristics, molecular subtypes, receptors statuses, and survival in a cohort of Croatian BC patients. Results showed that higher SHH and AR expressions were associated with positive receptor status, but increased SHH expression had a negative impact on survival in receptor-negative BCs. On the contrary, higher AR expression was mostly protective. However, multivariate analysis showed that only higher AR expression could be considered as an independent prognostic biomarker for poorer overall survival in triple-negative breast cancer patients (TNBC) (HR 10.9, 95% CI 1.43-83.67; p = 0.021), what could be Croatian population-related. SHH could be a potential target for treating TNBCs and HER2-enriched BCs, in cases where HH-GLI signaling is canonical (SHH-dependent).
Collapse
Affiliation(s)
- Ivan Budimir
- Department of Plastic, Reconstructive and Aesthetic Surgery, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Čedna Tomasović-Lončarić
- Clinical Department of Pathology and Cytology, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Kristina Kralik
- Department of Medical Statistics and Medical Informatics, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Josipa Čonkaš
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Domagoj Eljuga
- Department of Plastic, Reconstructive and Aesthetic Surgery, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Faculty of Health Sciences, Libertas International University, 10000 Zagreb, Croatia
| | - Rado Žic
- Department of Plastic, Reconstructive and Aesthetic Surgery, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Božo Gorjanc
- Department of Plastic, Reconstructive and Aesthetic Surgery, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Hrvoje Tucaković
- Department of Plastic, Reconstructive and Aesthetic Surgery, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Doroteja Caktaš
- Department of Plastic, Reconstructive and Aesthetic Surgery, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Josip Jaman
- Department of Plastic, Reconstructive and Aesthetic Surgery, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Valentino Lisek
- Department of Abdominal Surgery, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Zlatko Vlajčić
- Department of Plastic, Reconstructive and Aesthetic Surgery, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Krešimir Martić
- Department of Plastic, Reconstructive and Aesthetic Surgery, Dubrava University Hospital, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Correspondence: (K.M.); (P.O.); Tel.: +385-98-752-178 (K.M.); +385-98-659-083 (P.O.)
| | - Petar Ozretić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
- Correspondence: (K.M.); (P.O.); Tel.: +385-98-752-178 (K.M.); +385-98-659-083 (P.O.)
| |
Collapse
|
18
|
Cancer stem cell markers interplay with chemoresistance in triple negative breast cancer: A therapeutic perspective. Bull Cancer 2022; 109:960-971. [DOI: 10.1016/j.bulcan.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/18/2022] [Accepted: 05/03/2022] [Indexed: 11/19/2022]
|
19
|
Xu H, Zhang F, Gao X, Zhou Q, Zhu L. Fate decisions of breast cancer stem cells in cancer progression. Front Oncol 2022; 12:968306. [PMID: 36046046 PMCID: PMC9420991 DOI: 10.3389/fonc.2022.968306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Breast cancer has a marked recurrence and metastatic trait and is one of the most prevalent malignancies affecting women’s health worldwide. Tumor initiation and progression begin after the cell goes from a quiescent to an activated state and requires different mechanisms to act in concert to regulate t a specific set of spectral genes for expression. Cancer stem cells (CSCs) have been proven to initiate and drive tumorigenesis due to their capability of self-renew and differentiate. In addition, CSCs are believed to be capable of causing resistance to anti-tumor drugs, recurrence and metastasis. Therefore, exploring the origin, regulatory mechanisms and ultimate fate decision of CSCs in breast cancer outcomes has far-reaching clinical implications for the development of breast cancer stem cell (BCSC)-targeted therapeutic strategies. In this review, we will highlight the contribution of BCSCs to breast cancer and explore the internal and external factors that regulate the fate of BCSCs.
Collapse
|
20
|
Guha L, Bhat IA, Bashir A, Rahman JU, Pottoo FH. Nanotechnological Approaches for the Treatment of Triple-Negative Breast Cancer: A Comprehensive Review. Curr Drug Metab 2022; 23:781-799. [PMID: 35676850 DOI: 10.2174/1389200223666220608144551] [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: 12/03/2021] [Revised: 02/01/2022] [Accepted: 03/10/2022] [Indexed: 01/05/2023]
Abstract
Breast cancer is the most prevalent cancer in women around the world, having a sudden spread nowadays because of the poor sedentary lifestyle of people. Comprising several subtypes, one of the most dangerous and aggressive ones is triple-negative breast cancer or TNBC. Even though conventional surgical approaches like single and double mastectomy and preventive chemotherapeutic approaches are available, they are not selective to cancer cells and are only for symptomatic treatment. A new branch called nanotechnology has emerged in the last few decades that offers various novel characteristics, such as size in nanometric scale, enhanced adherence to multiple targeting moieties, active and passive targeting, controlled release, and site-specific targeting. Among various nanotherapeutic approaches like dendrimers, lipid-structured nanocarriers, carbon nanotubes, etc., nanoparticle targeted therapeutics can be termed the best among all for their specific cytotoxicity to cancer cells and increased bioavailability to a target site. This review focuses on the types and molecular pathways involving TNBC, existing treatment strategies, various nanotechnological approaches like exosomes, carbon nanotubes, dendrimers, lipid, and carbon-based nanocarriers, and especially various nanoparticles (NPs) like polymeric, photodynamic, peptide conjugated, antibody-conjugated, metallic, inorganic, natural product capped, and CRISPR based nanoparticles already approved for treatment or are under clinical and pre-clinical trials for TNBC.
Collapse
Affiliation(s)
- Lahanya Guha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Mohali, S.A.S Nagar, Punjab 160062, India
| | - Ishfaq Ahmad Bhat
- Northern Railway Hospital, Sri Mata Vaishno Devi, Katra, Reasi 182320, India
| | - Aasiya Bashir
- Department of Pharmaceutical Sciences, Faculty of Applied Sciences and Technology, University of Kashmir, Hazratbal, Srinagar-190006, J&K, India
| | - Jawad Ur Rahman
- Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam 31441, Saudi Arabia
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
21
|
Yamashita N, Kufe D. Addiction of Cancer Stem Cells to MUC1-C in Triple-Negative Breast Cancer Progression. Int J Mol Sci 2022; 23:8219. [PMID: 35897789 PMCID: PMC9331006 DOI: 10.3390/ijms23158219] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive malignancy with limited treatment options. TNBC progression is associated with expansion of cancer stem cells (CSCs). Few insights are available regarding druggable targets that drive the TNBC CSC state. This review summarizes the literature on TNBC CSCs and the compelling evidence that they are addicted to the MUC1-C transmembrane protein. In normal epithelia, MUC1-C is activated by loss of homeostasis and induces reversible wound-healing responses of inflammation and repair. However, in settings of chronic inflammation, MUC1-C promotes carcinogenesis. MUC1-C induces EMT, epigenetic reprogramming and chromatin remodeling in TNBC CSCs, which are dependent on MUC1-C for self-renewal and tumorigenicity. MUC1-C-induced lineage plasticity in TNBC CSCs confers DNA damage resistance and immune evasion by chronic activation of inflammatory pathways and global changes in chromatin architecture. Of therapeutic significance, an antibody generated against the MUC1-C extracellular domain has been advanced in a clinical trial of anti-MUC1-C CAR T cells and in IND-enabling studies for development as an antibody-drug conjugate (ADC). Agents targeting the MUC1-C cytoplasmic domain have also entered the clinic and are undergoing further development as candidates for advancing TNBC treatment. Eliminating TNBC CSCs will be necessary for curing this recalcitrant cancer and MUC1-C represents a promising druggable target for achieving that goal.
Collapse
Affiliation(s)
- Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
22
|
Scalable multiplex co-fractionation/mass spectrometry platform for accelerated protein interactome discovery. Nat Commun 2022; 13:4043. [PMID: 35831314 PMCID: PMC9279285 DOI: 10.1038/s41467-022-31809-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 06/29/2022] [Indexed: 12/14/2022] Open
Abstract
Co-fractionation/mass spectrometry (CF/MS) enables the mapping of endogenous macromolecular networks on a proteome scale, but current methods are experimentally laborious, resource intensive and afford lesser quantitative accuracy. Here, we present a technically efficient, cost-effective and reproducible multiplex CF/MS (mCF/MS) platform for measuring and comparing, simultaneously, multi-protein assemblies across different experimental samples at a rate that is up to an order of magnitude faster than previous approaches. We apply mCF/MS to map the protein interaction landscape of non-transformed mammary epithelia versus breast cancer cells in parallel, revealing large-scale differences in protein-protein interactions and the relative abundance of associated macromolecules connected with cancer-related pathways and altered cellular processes. The integration of multiplexing capability within an optimized workflow renders mCF/MS as a powerful tool for systematically exploring physical interaction networks in a comparative manner. Co-fractionation/mass spectrometry (CF/MS) allows mapping protein interactomes but efficiency and quantitative accuracy are limited. Here, the authors develop a reproducible multiplexed CF/MS method and apply it to characterize interactome rewiring in breast cancer cells.
Collapse
|
23
|
Pouliquen DL, Boissard A, Henry C, Coqueret O, Guette C. Curcuminoids as Modulators of EMT in Invasive Cancers: A Review of Molecular Targets With the Contribution of Malignant Mesothelioma Studies. Front Pharmacol 2022; 13:934534. [PMID: 35873564 PMCID: PMC9304619 DOI: 10.3389/fphar.2022.934534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/15/2022] [Indexed: 11/21/2022] Open
Abstract
Curcuminoids, which include natural acyclic diarylheptanoids and the synthetic analogs of curcumin, have considerable potential for fighting against all the characteristics of invasive cancers. The epithelial-to-mesenchymal transition (EMT) is a fundamental process for embryonic morphogenesis, however, the last decade has confirmed it orchestrates many features of cancer invasiveness, such as tumor cell stemness, metabolic rewiring, and drug resistance. A wealth of studies has revealed EMT in cancer is in fact driven by an increasing number of parameters, and thus understanding its complexity has now become a cornerstone for defining future therapeutic strategies dealing with cancer progression and metastasis. A specificity of curcuminoids is their ability to target multiple molecular targets, modulate several signaling pathways, modify tumor microenvironments and enhance the host’s immune response. Although the effects of curcumin on these various parameters have been the subject of many reviews, the role of curcuminoids against EMT in the context of cancer have never been reviewed so far. This review first provides an updated overview of all EMT drivers, including signaling pathways, transcription factors, non-coding RNAs (ncRNAs) and tumor microenvironment components, with a special focus on the most recent findings. Secondly, for each of these drivers the effects of curcumin/curcuminoids on specific molecular targets are analyzed. Finally, we address some common findings observed between data reported in the literature and the results of investigations we conducted on experimental malignant mesothelioma, a model of invasive cancer representing a useful tool for studies on EMT and cancer.
Collapse
Affiliation(s)
- Daniel L. Pouliquen
- Inserm, CNRS, Nantes Université, CRCI2NA, Université d’Angers, Angers, France
- *Correspondence: Daniel L. Pouliquen,
| | - Alice Boissard
- ICO, Inserm, CNRS, Nantes Université, CRCI2NA, Université d’Angers, Angers, France
| | - Cécile Henry
- ICO, Inserm, CNRS, Nantes Université, CRCI2NA, Université d’Angers, Angers, France
| | - Olivier Coqueret
- Inserm, CNRS, Nantes Université, CRCI2NA, Université d’Angers, Angers, France
| | - Catherine Guette
- ICO, Inserm, CNRS, Nantes Université, CRCI2NA, Université d’Angers, Angers, France
| |
Collapse
|
24
|
Dewi C, Fristiohady A, Amalia R, Khairul Ikram NK, Ibrahim S, Muchtaridi M. Signaling Pathways and Natural Compounds in Triple-Negative Breast Cancer Cell Line. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123661. [PMID: 35744786 PMCID: PMC9227697 DOI: 10.3390/molecules27123661] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, having a poor prognosis and rapid metastases. TNBC is characterized by the absence of estrogen, progesterone, and human epidermal growth receptor-2 (HER2) expressions and has a five-year survival rate. Compared to other breast cancer subtypes, TNBC patients only respond to conventional chemotherapies, and even then, with limited success. Shortages of chemotherapeutic medication can lead to resistance, pressured index therapy, non-selectivity, and severe adverse effects. Finding targeted treatments for TNBC is difficult owing to the various features of cancer. Hence, identifying the most effective molecular targets in TNBC pathogenesis is essential for predicting response to targeted therapies and preventing TNBC cell metastases. Nowadays, natural compounds have gained attention as TNBC treatments, and have offered new strategies for solving drug resistance. Here, we report a systematic review using the database from Pubmed, Science Direct, MDPI, BioScince, Springer, and Nature for articles screening from 2003 to 2022. This review analyzes relevant signaling pathways and the prospect of utilizing natural compounds as a therapeutic agent to improve TNBC treatments in the future.
Collapse
Affiliation(s)
- Citra Dewi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Pharmacy Department, Faculty of Science and Technology, Mandala Waluya University, Kendari 93561, Indonesia
| | - Adryan Fristiohady
- Faculty of Pharmacy, Halu Oleo University, Kampus Hijau Bumi Tridharma, Kendari 93232, Indonesia;
| | - Riezki Amalia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| | - Nur Kusaira Khairul Ikram
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Sugeng Ibrahim
- Department of Molecular Biology, Faculty of Medicine, Universitas Katolik Soegijapranata, Semarang 50234, Indonesia;
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Correspondence:
| |
Collapse
|
25
|
Kandel N, Wang C. Hedgehog Autoprocessing: From Structural Mechanisms to Drug Discovery. Front Mol Biosci 2022; 9:900560. [PMID: 35669560 PMCID: PMC9163320 DOI: 10.3389/fmolb.2022.900560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Hedgehog (Hh) signaling plays pivotal roles in embryonic development. In adults, Hh signaling is mostly turned off but its abnormal activation is involved in many types of cancer. Hh signaling is initiated by the Hh ligand, generated from the Hh precursor by a specialized autocatalytic process called Hh autoprocessing. The Hh precursor consists of an N-terminal signaling domain (HhN) and a C-terminal autoprocessing domain (HhC). During Hh autoprocessing, the precursor is cleaved between N- and C-terminal domain followed by the covalent ligation of cholesterol to the last residue of HhN, which subsequently leads to the generation of Hh ligand for Hh signaling. Hh autoprocessing is at the origin of canonical Hh signaling and precedes all downstream signaling events. Mutations in the catalytic residues in HhC can lead to congenital defects such as holoprosencephaly (HPE). The aim of this review is to provide an in-depth summary of the progresses and challenges towards an atomic level understanding of the structural mechanisms of Hh autoprocessing. We also discuss drug discovery efforts to inhibit Hh autoprocessing as a new direction in cancer therapy.
Collapse
Affiliation(s)
- Nabin Kandel
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Chunyu Wang
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- *Correspondence: Chunyu Wang,
| |
Collapse
|
26
|
|
27
|
Chaudhuri A, Kumar DN, Dehari D, Singh S, Kumar P, Bolla PK, Kumar D, Agrawal AK. Emergence of Nanotechnology as a Powerful Cavalry against Triple-Negative Breast Cancer (TNBC). Pharmaceuticals (Basel) 2022; 15:542. [PMID: 35631368 PMCID: PMC9143332 DOI: 10.3390/ph15050542] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is considered one of the un-manageable types of breast cancer, involving devoid of estrogen, progesterone, and human epidermal growth factor receptor 2 (HER 2) receptors. Due to their ability of recurrence and metastasis, the management of TNBC remains a mainstay challenge, despite the advancements in cancer therapies. Conventional chemotherapy remains the only treatment regimen against TNBC and suffers several limitations such as low bioavailability, systemic toxicity, less targetability, and multi-drug resistance. Although various targeted therapies have been introduced to manage the hardship of TNBC, they still experience certain limitations associated with the survival benefits. The current research thus aimed at developing and improving the strategies for effective therapy against TNBC. Such strategies involved the emergence of nanoparticles. Nanoparticles are designated as nanocavalries, loaded with various agents (drugs, genes, etc.) to battle the progression and metastasis of TNBC along with overcoming the limitations experienced by conventional chemotherapy and targeted therapy. This article documents the treatment regimens of TNBC along with their efficacy towards different subtypes of TNBC, and the various nanotechnologies employed to increase the therapeutic outcome of FDA-approved drug regimens.
Collapse
Affiliation(s)
- Aiswarya Chaudhuri
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (A.C.); (D.N.K.); (D.D.); (S.S.); (D.K.)
| | - Dulla Naveen Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (A.C.); (D.N.K.); (D.D.); (S.S.); (D.K.)
| | - Deepa Dehari
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (A.C.); (D.N.K.); (D.D.); (S.S.); (D.K.)
| | - Sanjay Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (A.C.); (D.N.K.); (D.D.); (S.S.); (D.K.)
- Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India
| | - Pradeep Kumar
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
| | - Pradeep Kumar Bolla
- Department of Biomedical Engineering, College of Engineering, The University of Texas at El Paso, 500 W. University Ave, El Paso, TX 79968, USA;
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (A.C.); (D.N.K.); (D.D.); (S.S.); (D.K.)
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (A.C.); (D.N.K.); (D.D.); (S.S.); (D.K.)
| |
Collapse
|
28
|
Shrihastini V, Muthuramalingam P, Adarshan S, Sujitha M, Chen JT, Shin H, Ramesh M. Plant Derived Bioactive Compounds, Their Anti-Cancer Effects and In Silico Approaches as an Alternative Target Treatment Strategy for Breast Cancer: An Updated Overview. Cancers (Basel) 2021; 13:cancers13246222. [PMID: 34944840 PMCID: PMC8699774 DOI: 10.3390/cancers13246222] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer is one of the most common malignant diseases that occur worldwide, among which breast cancer is the second leading cause of death in women. The subtypes are associated with differences in the outcome and were selected for treatments according to the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor. Triple-negative breast cancer, one of the subtypes of breast cancer, is difficult to treat and can even lead to death. If breast cancer is not treated during the initial stages, it may spread to nearby organs, a process called metastasis, through the blood or lymph system. For in vitro studies, MCF-7, MDA-MB-231, MDA-MB-468, and T47B are the most commonly used breast cancer cell lines. Clinically, chemotherapy and radiotherapy are usually expensive and can also cause side effects. To overcome these issues, medicinal plants could be the best alternative for chemotherapeutic drugs with fewer side effects and cost-effectiveness. Furthermore, the genes involved in breast cancer can be regulated and synergized with signaling molecules to suppress the proliferation of breast cancer cells. In addition, nanoparticles encapsulating (nano-encapsulation) medicinal plant extracts showed a significant reduction in the apoptotic and cytotoxic activities of breast cancer cells. This present review mainly speculates an overview of the native medicinal plant derived anti-cancerous compounds with its efficiency, types and pathways involved in breast cancer along with its genes, the mechanism of breast cancer brain metastasis, chemoresistivity and its mechanism, bioinformatics approaches which could be an effective alternative for drug discovery.
Collapse
Affiliation(s)
- Vijayakumar Shrihastini
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore 641062, Tamil Nadu, India; (V.S.); (M.S.)
| | - Pandiyan Muthuramalingam
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore 641062, Tamil Nadu, India; (V.S.); (M.S.)
- Correspondence: (P.M.); (J.-T.C.)
| | - Sivakumar Adarshan
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi 630003, Tamil Nadu, India; (S.A.); (M.R.)
| | - Mariappan Sujitha
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore 641062, Tamil Nadu, India; (V.S.); (M.S.)
| | - Jen-Tsung Chen
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung 811, Taiwan
- Correspondence: (P.M.); (J.-T.C.)
| | - Hyunsuk Shin
- Department of Horticultural Sciences, Gyeongsang National University, Jinju 52725, Korea;
| | - Manikandan Ramesh
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi 630003, Tamil Nadu, India; (S.A.); (M.R.)
| |
Collapse
|
29
|
He L, Wick N, Germans SK, Peng Y. The Role of Breast Cancer Stem Cells in Chemoresistance and Metastasis in Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:cancers13246209. [PMID: 34944829 PMCID: PMC8699562 DOI: 10.3390/cancers13246209] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 02/05/2023] Open
Abstract
Triple negative breast cancer (TNBC) remains an aggressive disease due to the lack of targeted therapies and low rate of response to chemotherapy that is currently the main treatment modality for TNBC. Breast cancer stem cells (BCSCs) are a small subpopulation of breast tumors and recognized as drivers of tumorigenesis. TNBC tumors are characterized as being enriched for BCSCs. Studies have demonstrated the role of BCSCs as the source of metastatic disease and chemoresistance in TNBC. Multiple targets against BCSCs are now under investigation, with the considerations of either selectively targeting BCSCs or co-targeting BCSCs and non-BCSCs (majority of tumor cells). This review article provides a comprehensive overview of recent advances in the role of BCSCs in TNBC and the identification of cancer stem cell biomarkers, paving the way for the development of new targeted therapies. The review also highlights the resultant discovery of cancer stem cell targets in TNBC and the ongoing clinical trials treating chemoresistant breast cancer. We aim to provide insights into better understanding the mutational landscape of BCSCs and exploring potential molecular signaling pathways targeting BCSCs to overcome chemoresistance and prevent metastasis in TNBC, ultimately to improve the overall survival of patients with this devastating disease.
Collapse
Affiliation(s)
- Lin He
- Department of Pathology, University of Texas Southwestern Medical Center, 6201 Harry Hines Blvd, Dallas, TX 75235, USA; (L.H.); (N.W.); (S.K.G.)
| | - Neda Wick
- Department of Pathology, University of Texas Southwestern Medical Center, 6201 Harry Hines Blvd, Dallas, TX 75235, USA; (L.H.); (N.W.); (S.K.G.)
| | - Sharon Koorse Germans
- Department of Pathology, University of Texas Southwestern Medical Center, 6201 Harry Hines Blvd, Dallas, TX 75235, USA; (L.H.); (N.W.); (S.K.G.)
| | - Yan Peng
- Department of Pathology, University of Texas Southwestern Medical Center, 6201 Harry Hines Blvd, Dallas, TX 75235, USA; (L.H.); (N.W.); (S.K.G.)
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75235, USA
- Correspondence:
| |
Collapse
|
30
|
Hedgehog Pathway Inhibitors against Tumor Microenvironment. Cells 2021; 10:cells10113135. [PMID: 34831357 PMCID: PMC8619966 DOI: 10.3390/cells10113135] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/24/2022] Open
Abstract
Targeting the hedgehog (HH) pathway to treat aggressive cancers of the brain, breast, pancreas, and prostate has been ongoing for decades. Gli gene amplifications have been long discovered within malignant glioma patients, and since then, inhibitors against HH pathway-associated molecules have successfully reached the clinical stage where several of them have been approved by the FDA. Albeit this success rate implies suitable progress, clinically used HH pathway inhibitors fail to treat patients with metastatic or recurrent disease. This is mainly due to heterogeneous tumor cells that have acquired resistance to the inhibitors along with the obstacle of effectively targeting the tumor microenvironment (TME). Severe side effects such as hyponatremia, diarrhea, fatigue, amenorrhea, nausea, hair loss, abnormal taste, and weight loss have also been reported. Furthermore, HH signaling is known to be involved in the regulation of immune cell maturation, angiogenesis, inflammation, and polarization of macrophages and myeloid-derived suppressor cells. It is critical to determine key mechanisms that can be targeted at different levels of tumor development and progression to address various clinical issues. Hence current research focus encompasses understanding how HH controls TME to develop TME altering and combinatorial targeting strategies. In this review, we aim to discuss the pros and cons of targeting HH signaling molecules, understand the mechanism involved in treatment resistance, reveal the role of the HH pathway in anti-tumor immune response, and explore the development of potential combination treatment of immune checkpoint inhibitors with HH pathway inhibitors to target HH-driven cancers.
Collapse
|
31
|
Mani C, Tripathi K, Omy TR, Reedy M, Manne U, Palle K. GLI1-targeting drugs induce replication stress and homologous recombination deficiency and synergize with PARP-targeted therapies in triple negative breast cancer cells. Biochim Biophys Acta Mol Basis Dis 2021; 1868:166300. [PMID: 34748904 DOI: 10.1016/j.bbadis.2021.166300] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/20/2021] [Accepted: 11/03/2021] [Indexed: 01/20/2023]
Abstract
Triple negative breast cancer (TNBC), an aggressive and highly metastatic subtype of breast cancer. Glioma-associated oncogene 1 (GLI1) is a transcription factor and effector of the Hedgehog (Hh) signaling pathway, and is predictive of poor survival for TNBC patients. A nanostring DNA Damage Response (DDR) mRNA panel was used to identify GLI1-induced regulation of DDR genes. Western blots, immunohistochemistry and immunofluorescence were used to evaluate protein expression. Colony assays and mammosphere formation assays were utilized to assess survival of cancer cells. Flow cytometry analyses were employed to evaluate changes in the cell cycle profile, and DNA fiber assays were used to analyze alterations in replication dynamics in TNBC cells. The UALCAN portal and Ensemble programs were used for computational analysis of TCGA data. CompuSyn software was used to calculate combination index (CI) values to assess synergism in drug combination experiments. Inhibition of GLI1 in TNBC cells transcriptionally downregulate expression of FANCD2 and its foci formation, and causes a homologous recombination repair (HR) deficiency. As HR-deficient cancer cells are sensitive to PARP-targeted therapies, we evaluated a combination of the GLI1 inhibitor, GANT61, and a PARP inhibitor (olaparib) in TNBC cells. Combination of GANT61 and olaparib elevated DNA damage levels and these drug combinations caused synergistic lethality to TNBC cells. Aberrantly activated GLI1 regulates HR-mediated DNA repair by transcriptionally regulating FANCD2 to overcome chemotherapy-induced replication stress and DNA damage, and it contributes to resistance of TNBC cells to therapeutics.
Collapse
Affiliation(s)
- Chinnadurai Mani
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Kaushlendra Tripathi
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36904, USA
| | - Tasmin R Omy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Mark Reedy
- Department of Obstetrics and Gynecology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Komaraiah Palle
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
32
|
Garcia N, Ulin M, Ali M, Al-Hendy A, Carvalho KC, Yang Q. Evaluation of Hedgehog Pathway Inhibitors as a Therapeutic Option for Uterine Leiomyosarcoma Using the Xenograft Model. Reprod Sci 2021; 29:781-790. [PMID: 34642915 PMCID: PMC8863774 DOI: 10.1007/s43032-021-00731-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 08/26/2021] [Indexed: 01/20/2023]
Abstract
Uterine leiomyosarcoma (LMS) contributes to a significant proportion of uterine cancer deaths. It is a rare and high-risk gynecological cancer. LMS is challenging to the treatment due to the resistance of several therapies. The activation of the Hedgehog (HH) pathway has been reported in several types of female cancers. Uterine LMS presents an upregulation of the crucial HH signaling pathway members such as SMO and GLI1. Although targeting the HH pathway exhibited a potent inhibitory effect on the phenotype of uterine LMS in vitro, the effect of the HH inhibitors on LMS growth in vivo has not been identified. The present study aimed to assess the effect of Hedgehog pathway inhibitors (SMO-LDE225 and GLI-Gant61) as a therapeutic option in the xenograft model of uterine LMS. The results demonstrated that LDE225 treatment did not show any inhibitory effect on LMS tumor growth; however, treatment with GLI inhibitor (Gant61) induced a remarkable tumor regression with a significant decrease in Ki67 expression, compared to control (p < 0.01). Moreover, administration of Gant61 decreased the expression of GLI1, GLI target genes BMP4 and c-MYC (p < 0.05), indicating that the HH pathway is implicated in the LMS experimental model. In conclusion, our studies demonstrate for the first time that GLI inhibitor (Gant61), but not SMO inhibitor (LDE225), shows a potent inhibitory effect on LMS tumor growth and concomitantly suppresses the expression of GLI1- and GLI-targeted genes using the xenograft model of uterine LMS.
Collapse
Affiliation(s)
- Natalia Garcia
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA.,Laboratório de Ginecologia Estrutural e Molecular - LIM 58, Disciplina de Ginecologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, HCFMUSP, Sao Paulo, Brazil
| | - Mara Ulin
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Mohamed Ali
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA.,Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - Katia Candido Carvalho
- Laboratório de Ginecologia Estrutural e Molecular - LIM 58, Disciplina de Ginecologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, HCFMUSP, Sao Paulo, Brazil
| | - Qiwei Yang
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
33
|
Astarita EM, Maloney SM, Hoover CA, Berkeley BJ, VanKlompenberg MK, Nair TM, Prosperi JR. Adenomatous Polyposis Coli loss controls cell cycle regulators and response to paclitaxel in MDA-MB-157 metaplastic breast cancer cells. PLoS One 2021; 16:e0255738. [PMID: 34370741 PMCID: PMC8351968 DOI: 10.1371/journal.pone.0255738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/22/2021] [Indexed: 11/22/2022] Open
Abstract
Adenomatous Polyposis Coli (APC) is lost in approximately 70% of sporadic breast cancers, with an inclination towards triple negative breast cancer (TNBC). TNBC is treated with traditional chemotherapy, such as paclitaxel (PTX); however, tumors often develop drug resistance. We previously created APC knockdown cells (APC shRNA1) using the human TNBC cells, MDA-MB-157, and showed that APC loss induces PTX resistance. To understand the mechanisms behind APC-mediated PTX response, we performed cell cycle analysis and analyzed cell cycle related proteins. Cell cycle analysis indicated increased G2/M population in both PTX-treated APC shRNA1 and parental cells, suggesting that APC expression does not alter PTX-induced G2/M arrest. We further studied the subcellular localization of the G2/M transition proteins, cyclin B1 and CDK1. The APC shRNA1 cells had increased CDK1, which was preferentially localized to the cytoplasm, and increased baseline CDK6. RNA-sequencing was performed to gain a global understanding of changes downstream of APC loss and identified a broad mis-regulation of cell cycle-related genes in APC shRNA1 cells. Our studies are the first to show an interaction between APC and taxane response in breast cancer. The implications include designing combination therapy to re-sensitize APC-mutant breast cancers to taxanes using the specific cell cycle alterations.
Collapse
Affiliation(s)
- Emily M. Astarita
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Chemistry/Biochemistry, University of Notre Dame, Notre Dame, IN, United States of America
| | - Sara M. Maloney
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, South Bend, IN, United States of America
| | - Camden A. Hoover
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
| | | | - Monica K. VanKlompenberg
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, South Bend, IN, United States of America
| | - T. Murlidharan Nair
- Department of Biology and Computer Science/Informatics, Indiana University South Bend, South Bend, IN, United States of America
| | - Jenifer R. Prosperi
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, South Bend, IN, United States of America
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
| |
Collapse
|
34
|
Jiang C, Wang X, Teng B, Wang Z, Li F, Zhao Y, Guo Y, Zeng Q. Peptide-Targeted High-Density Lipoprotein Nanoparticles for Combinatorial Treatment against Metastatic Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2021; 13:35248-35265. [PMID: 34284582 DOI: 10.1021/acsami.1c02074] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The sonic hedgehog (SHH) signaling pathway exhibits aberrant activation in triple-negative breast cancer (TNBC), wherein it regulates several malignant phenotypes related to tumor metastasis. GANT61, an inhibitor of the SHH signaling pathway, may offer promise when administered in combination with conventional chemotherapy to treat metastatic TNBC. However, poor bioavailability and substantial off-target toxicity limit its clinical application. To address these limitations, we designed a peptide-functionalized dual-targeting delivery system encapsulating paclitaxel and GANT61 in tLyP-1 peptide-modified reconstituted high-density lipoprotein nanoparticle (tLyP-1-rHDL-PTX/GANT61 NP) for metastatic TNBC treatment. The apolipoprotein A-1 and tLyP-1 peptide modified on the surface of nanoparticles enable the delivery system to target tumor cells by binding to the overexpressed scavenger receptor B type I and neuropilin-1 receptor. Moreover, the tLyP-1 peptide also enables the deep tumor penetration of nanoparticles further facilitating paclitaxel and GANT61 delivery. Increased cellular uptake of the nanoparticles was observed in both MDA-MB-231, BT-549 tumor cells, and their 3D tumor spheroids. A series of in vitro experiments reveal that GANT61 was able to suppress key metastasis-related tumor cell activities including angiogenesis, migration, invasion, and stemness. Owing to more effective drug administration, the metastasis suppression efficiency of GANT61 was significantly enhanced by the dual-targeting tLyP-1-rHDL delivery system. Meanwhile, the codelivery of paclitaxel and GANT61 by dual-targeting tLyP-1-rHDL nanoparticles demonstrated superior efficiency of disrupting proliferation and inducing apoptosis in tumor cells compared with drug solutions. In a spontaneous metastasis breast cancer NCG mice model, the tLyP-1-rHDL-PTX/GANT61 nanoparticles exhibited highly tumor-specific distribution and result in significant inhibition of the primary tumor growth and dramatic reduction of lung metastasis without obvious side effects. The present work suggests that a combination of the SHH signaling pathway suppression and chemotherapy assisted by peptide-functionalized targeting tLyP-1-rHDL nanoparticles may provide a promising strategy for metastatic TNBC treatment.
Collapse
Affiliation(s)
- Chuli Jiang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xingyue Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Biyun Teng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhe Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fenghe Li
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yu Zhao
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuan Guo
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qiu Zeng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
35
|
Qayoom H, Wani NA, Alshehri B, Mir MA. An insight into the cancer stem cell survival pathways involved in chemoresistance in triple-negative breast cancer. Future Oncol 2021; 17:4185-4206. [PMID: 34342489 DOI: 10.2217/fon-2021-0172] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most complex, aggressive and fatal subtype of breast cancer. Owing to the lack of targeted therapy and heterogenic nature of TNBC, chemotherapy remains the sole treatment option for TNBC, with taxanes and anthracyclines representing the general chemotherapeutic regimen in TNBC therapy. But unfortunately, patients develop resistance to the existing chemotherapeutic regimen, resulting in approximately 90% treatment failure. Breast cancer stem cells (BCSCs) are one of the major causes for the development of chemoresistance in TNBC patients. After surviving the chemotherapy damage, the presence of BCSCs results in relapse and recurrence of TNBC. Several pathways are known to regulate BCSCs' survival, such as the Wnt/β-catenin, Hedgehog, JAK/STAT and HIPPO pathways. Therefore it is imperative to target these pathways in the context of eliminating chemoresistance. In this review we will discuss the novel strategies and various preclinical and clinical studies to give an insight into overcoming TNBC chemoresistance. We present a detailed account of recent studies carried out that open an exciting perspective in relation to the mechanisms of chemoresistance.
Collapse
Affiliation(s)
- Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, J&K, India
| | - Nissar A Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir Nunar Ganderbal 191201, J&K, India
| | - Bader Alshehri
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, KSA
| | - Manzoor A Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, J&K, India
| |
Collapse
|
36
|
Fultang N, Chakraborty M, Peethambaran B. Regulation of cancer stem cells in triple negative breast cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:321-342. [PMID: 35582030 PMCID: PMC9019272 DOI: 10.20517/cdr.2020.106] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/28/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
Triple Negative Breast Cancer (TNBC) is the most lethal subtype of breast cancer. Despite the successes of emerging targeted therapies, relapse, recurrence, and therapy failure rates in TNBC significantly outpace other subtypes of breast cancer. Mounting evidence suggests accumulation of therapy resistant Cancer Stem Cell (CSC) populations within TNBCs contributes to poor clinical outcomes. These CSCs are enriched in TNBC compared to non-TNBC breast cancers. The mechanisms underlying CSC accumulation have been well-characterized and discussed in other reviews. In this review, we focus on TNBC-specific mechanisms that allow the expansion and activity of self-renewing CSCs. We highlight cellular signaling pathways and transcription factors, specifically enriched in TNBC over non-TNBC breast cancer, contributing to stemness. We also analyze publicly available single-cell RNA-seq data from basal breast cancer tumors to highlight the potential of emerging bioinformatic approaches in identifying novel drivers of stemness in TNBC and other cancers.
Collapse
Affiliation(s)
- Norman Fultang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19140, USA
| | - Madhuparna Chakraborty
- Department of Biological Sciences, The University of the Sciences, Philadelphia, PA 19140, USA
| | - Bela Peethambaran
- Department of Biological Sciences, The University of the Sciences, Philadelphia, PA 19140, USA
| |
Collapse
|
37
|
Differential Expression of BOC, SPOCK2, and GJD3 Is Associated with Brain Metastasis of ER-Negative Breast Cancers. Cancers (Basel) 2021; 13:cancers13122982. [PMID: 34203581 PMCID: PMC8232218 DOI: 10.3390/cancers13122982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Brain metastasis is diagnosed in 30–50% of metastatic breast cancer patients with currently limited treatment strategies and usually short survival rates. In the present study, we aim to identify genes specifically associated with the development of brain metastasis in breast cancer. Therefore, we compared RNA expression profiles from two groups of patients with metastatic breast cancer, with and without brain involvement. Three genes BOC, SPOCK2, and GJD3 were overexpressed in the group of primary breast cancers which developed brain metastasis. Expression profiles were confirmed in an independent breast cancer cohort for both BOC and SPOCK2. In addition, differential overexpression of SPOCK2 and GJD3 mRNA levels were found to be associated with the development of brain metastasis in an external online database of 204 primary breast cancers. Verification of these genes as biomarkers for brain metastasis development in primary breast cancer is warranted. Abstract Background: Brain metastasis is considered one of the major causes of mortality in breast cancer patients. To invade the brain, tumor cells need to pass the blood-brain barrier by mechanisms that are partially understood. In primary ER-negative breast cancers that developed brain metastases, we found that some of the differentially expressed genes play roles in the T cell response. The present study aimed to identify genes involved in the formation of brain metastasis independently from the T cell response. Method: Previously profiled primary breast cancer samples were reanalyzed. Genes that were found to be differentially expressed were confirmed by RT-PCR and by immunohistochemistry using an independent cohort of samples. Results: BOC, SPOCK2, and GJD3 were overexpressed in the primary breast tumors that developed brain metastasis. BOC expression was successfully validated at the protein level. SPOCK2 was validated at both mRNA and protein levels. SPOCK2 and GJD3 mRNA overexpression were also found to be associated with cerebral metastasis in an external online database consisting of 204 primary breast cancers. Conclusion: The overexpression of BOC, SPOCK2, and GJD3 is associated with the invasion of breast cancer into the brain. Further studies to determine their specific function and potential value as brain metastasis biomarkers are required.
Collapse
|
38
|
Soheilifar MH, Masoudi-Khoram N, Madadi S, Nobari S, Maadi H, Keshmiri Neghab H, Amini R, Pishnamazi M. Angioregulatory microRNAs in breast cancer: Molecular mechanistic basis and implications for therapeutic strategies. J Adv Res 2021; 37:235-253. [PMID: 35499045 PMCID: PMC9039675 DOI: 10.1016/j.jare.2021.06.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/13/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer-associated angiogenesis is a fundamental process in tumor growth and metastasis. Angioregulatory miRNA–target gene interaction is not only involved in sprouting vessels of breast tumors but also, trans-differentiation of breast cancer cells to endothelial cells in a process termed vasculogenic mimicry. Successful targeting of tumor angiogenesis is still a missing link in the treatment of Breast cancer (BC) due to the low effectiveness of anti-angiogenic therapies in this cancer. Response to anti-angiogenic therapeutics are controlled by a miRNAs, so the identification of interaction networks of miRNAs–targets can be applicable in determining anti-angiogeneic therapy and new biomarkers in BC. Angioregulatory miRNAs in breast cancer cells and their microenvironment have therapeutic potential in cancer treatment.
Background Cancer-associated angiogenesis is a fundamental process in tumor growth and metastasis. A variety of signaling regulators and pathways contribute to establish neovascularization, among them as small endogenous non-coding RNAs, microRNAs (miRNAs) play prominent dual regulatory function in breast cancer (BC) angiogenesis. Aim of Review This review aims at describing the current state-of-the-art in BC angiogenesis-mediated by angioregulatory miRNAs, and an overview of miRNAs dysregulation association with the anti-angiogenic response in addition to potential clinical application of miRNAs-based therapeutics. Key Scientific Concepts of Review Angioregulatory miRNA–target gene interaction is not only involved in sprouting vessels of breast tumors but also, trans-differentiation of BC cells to endothelial cells (ECs) in a process termed vasculogenic mimicry. Using canonical and non-canonical angiogenesis pathways, the tumor cell employs the oncogenic characteristics such as miRNAs dysregulation to increase survival, proliferation, oxygen and nutrient supply, and treatment resistance. Angioregulatory miRNAs in BC cells and their microenvironment have therapeutic potential in cancer treatment. Although, miRNAs dysregulation can serve as tumor biomarker nevertheless, due to the association of miRNAs dysregulation with anti-angiogenic resistant phenotype, clinical benefits of anti-angiogenic therapy might be challenging in BC. Hence, unveiling the molecular mechanism underlying angioregulatory miRNAs sparked a booming interest in finding new treatment strategies such as miRNA-based therapies in BC.
Collapse
Affiliation(s)
- Mohammad Hasan Soheilifar
- Department of Medical Laser, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Corresponding authorsat: Yara Institute, Academic Center for Education, Culture and Research (ACECR), Enghelab St, Tehran 1315795613, Iran (Mohammad Hasan Soheilifar). University of Limerick, Limerick V94 T9PX, Ireland (Mahboubeh Pishnamazi).
| | - Nastaran Masoudi-Khoram
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Soheil Madadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sima Nobari
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Maadi
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Hoda Keshmiri Neghab
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Razieh Amini
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahboubeh Pishnamazi
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Limerick, Ireland
- Corresponding authorsat: Yara Institute, Academic Center for Education, Culture and Research (ACECR), Enghelab St, Tehran 1315795613, Iran (Mohammad Hasan Soheilifar). University of Limerick, Limerick V94 T9PX, Ireland (Mahboubeh Pishnamazi).
| |
Collapse
|
39
|
García-Martínez A, Pérez-Balaguer A, Ortiz-Martínez F, Pomares-Navarro E, Sanmartín E, García-Escolano M, Montoyo-Pujol YG, Castellón-Molla E, Peiró G. Hedgehog gene expression patterns among intrinsic subtypes of breast cancer: Prognostic relevance. Pathol Res Pract 2021; 223:153478. [PMID: 34022683 DOI: 10.1016/j.prp.2021.153478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Hedgehog (Hh) signaling is a crucial developmental regulatory pathway recognized as a primary oncogenesis driver in various human cancers. However, its role in breast carcinoma (BC) has been underexplored. METHODS We analyzed the expression of several Hh associated genes in a clinical series and breast cancer cell lines. We included 193 BC stratified according to intrinsic immunophenotypes. Gene expression profiling ofBOC, PTCH, SMO, GLI1, GLI2, and GLI3 was performed by qRT-PCR. Results were correlated with clinical-pathological variables and outcome. RESULTS We observed expression ofGLI2 in triple-negative/basal-like (TN/BL) and GLI3 in luminal cells. In samples, BOC, GLI1, GLI2, and GLI3 expression correlated significantly with luminal tumors and good prognostic factors. In contrast, PTCH and SMO correlated with TN/BL phenotype and nodal involvement. Patients whose tumors expressed SMO had a poorer outcome, especially those with HER2 phenotype. Positive lymph-node status and high SMO remained independent poor prognostic factors. CONCLUSION Our results support a differential Hh pathway activation in BC phenotypes.SMO levels stratified patients at risk of recurrence and death in HER2 phenotype, and it showed an independent prognostic value. Therefore, SMO could be a potential therapeutic target for a subset of BC patients.
Collapse
Affiliation(s)
- Araceli García-Martínez
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain.
| | - Ariadna Pérez-Balaguer
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Fernando Ortiz-Martínez
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Eloy Pomares-Navarro
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Elena Sanmartín
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Marta García-Escolano
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Yoel G Montoyo-Pujol
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Elena Castellón-Molla
- Pathology Dept., University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Gloria Peiró
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain; Pathology Dept., University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| |
Collapse
|
40
|
Song K, Farzaneh M. Signaling pathways governing breast cancer stem cells behavior. Stem Cell Res Ther 2021; 12:245. [PMID: 33863385 PMCID: PMC8052733 DOI: 10.1186/s13287-021-02321-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the second common cancer and the leading cause of malignancy among females overall. Breast cancer stem cells (BCSCs) are a small population of breast cancer cells that play a critical role in the metastasis of breast cancer to other organs in the body. BCSCs have both self-renewal and differentiation capacities, which are thought to contribute to the aggressiveness of metastatic lesions. Therefore, targeting BCSCs can be a suitable approach for the treatment and metastasis of breast cancer. Growing evidence has indicated that the Wnt, NFκB, Notch, BMP2, STAT3, and hedgehog (Hh) signaling pathways govern epithelial-to-mesenchymal transition (EMT) activation, growth, and tumorigenesis of BCSCs in the primary regions. miRNAs as the central regulatory molecules also play critical roles in BCSC self-renewal, metastasis, and drug resistance. Hence, targeting these pathways might be a novel therapeutic approach for breast cancer diagnosis and therapy. This review discusses known signaling mechanisms involved in the stimulation or prevention of BCSC self-renewal, metastasis, and tumorigenesis.
Collapse
Affiliation(s)
- Kai Song
- Xuzhou Vocational College of Bioengineering, Xuzhou, 221006, Jiangsu, China.
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
41
|
Iriana S, Asha K, Repak M, Sharma-Walia N. Hedgehog Signaling: Implications in Cancers and Viral Infections. Int J Mol Sci 2021; 22:1042. [PMID: 33494284 PMCID: PMC7864517 DOI: 10.3390/ijms22031042] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022] Open
Abstract
The hedgehog (SHH) signaling pathway is primarily involved in embryonic gut development, smooth muscle differentiation, cell proliferation, adult tissue homeostasis, tissue repair following injury, and tissue polarity during the development of vertebrate and invertebrate organisms. GLIoma-associated oncogene homolog (GLI) family of zinc-finger transcription factors and smoothened (SMO) are the signal transducers of the SHH pathway. Both SHH ligand-dependent and independent mechanisms activate GLI proteins. Various transcriptional mechanisms, posttranslational modifications (phosphorylation, ubiquitination, proteolytic processing, SUMOylation, and acetylation), and nuclear-cytoplasmic shuttling control the activity of SHH signaling pathway proteins. The dysregulated SHH pathway is associated with bone and soft tissue sarcomas, GLIomas, medulloblastomas, leukemias, and tumors of breast, lung, skin, prostate, brain, gastric, and pancreas. While extensively studied in development and sarcomas, GLI family proteins play an essential role in many host-pathogen interactions, including bacterial and viral infections and their associated cancers. Viruses hijack host GLI family transcription factors and their downstream signaling cascades to enhance the viral gene transcription required for replication and pathogenesis. In this review, we discuss a distinct role(s) of GLI proteins in the process of tumorigenesis and host-pathogen interactions in the context of viral infection-associated malignancies and cancers due to other causes. Here, we emphasize the potential of the Hedgehog (HH) pathway targeting as a potential anti-cancer therapeutic approach, which in the future could also be tested in infection-associated fatalities.
Collapse
|
42
|
Targeting Hedgehog Pathway and DNA Methyltransferases in Uterine Leiomyosarcoma Cells. Cells 2020; 10:cells10010053. [PMID: 33396427 PMCID: PMC7824187 DOI: 10.3390/cells10010053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022] Open
Abstract
Uterine leiomyosarcoma (LMS) is an aggressive tumor that presents poor prognosis, high rates of recurrence and metastasis. Because of its rarity, there is no information available concerning LMS molecular mechanisms of origin and development. Here, we assessed the expression profile of Hedgehog (HH) signaling pathway markers and the effects of their pharmacological inhibition on uterine smooth muscle (UTSM), leiomyoma and LMS cells. Additionally, we also evaluated the effects of DNMTs inhibition on LMS cells behavior. Cell proliferation, migration and apoptosis rates were evaluated by MTT, Scratch and Annexin V assays, respectively. RNA expression and protein levels were assessed by qRT-PCR and Western blot. We found that SMO and GLIs (1, 2 and 3) expression was upregulated in LMS cells, with increased nuclear levels of GLI proteins. Treatment with LDE225 (SMOi) and Gant61 (GLIi) resulted in a significant reduction in Glis protein levels in LMS (p < 0.05). Additionally, the expression of DNMT (1, 3a, and 3b), as well as GLI1 nuclear expression, was significantly decreased after treatment with HH inhibitor in LMS cells. Our results showed that blocking of SMO, GLI and DNMTs is able to inhibit LMS proliferation, migration and invasion. Importantly, the combination of those treatments exhibited a potentiated effect on LMS malignant features due to HH pathway deactivation.
Collapse
|
43
|
Wang WJ, Wang H, Wang MS, Huang YQ, Ma YY, Qi J, Shi JP, Li W. Assessing the prognostic value of stemness-related genes in breast cancer patients. Sci Rep 2020; 10:18325. [PMID: 33110086 PMCID: PMC7591576 DOI: 10.1038/s41598-020-73164-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 08/14/2020] [Indexed: 01/21/2023] Open
Abstract
Breast cancer (BC) is currently one of the deadliest tumors worldwide. Cancer stem cells (CSCs) are a small group of tumor cells with self-renewal and differentiation abilities and high treatment resistance. One of the reasons for treatment failures is the inability to completely eliminate tumor stem cells. By using the edgeR package, we identified stemness-related differentially expressed genes in GSE69280. Via Lasso-penalized Cox regression analysis and univariate Cox regression analysis, survival genes were screened out to construct a prognostic model. Via nomograms and ROC curves, we verified the accuracy of the prognostic model. We selected 4 genes (PSMB9, CXCL13, NPR3, and CDKN2C) to establish a prognostic model from TCGA data and a validation model from GSE24450 data. We found that the low-risk score group had better OS than the high-risk score group, whether using TCGA or GSE24450 data. A prognostic model including four stemness-related genes was constructed in our study to determine targets of breast cancer stem cells (BCSCs) and improve the treatment effect.
Collapse
Affiliation(s)
- Wen-Jie Wang
- Department of Radio-Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, 26 Daoqian Street, Suzhou, 215001, Jiangsu, People's Republic of China
| | - Han Wang
- Department of Oncology, Jining Tumor Hospital, Jining, 272000, Shandong, People's Republic of China
| | - Meng-Sen Wang
- Department of Oncology, Jining No. 1 People's Hospital, Jining, 272011, Shandong, People's Republic of China
| | - Yue-Qing Huang
- Department of General Practice, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, People's Republic of China
| | - Yu-Yuan Ma
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, Jiangsu, People's Republic of China
| | - Jie Qi
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, Jiangsu, People's Republic of China
| | - Jian-Ping Shi
- Department of Radio-Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, 26 Daoqian Street, Suzhou, 215001, Jiangsu, People's Republic of China.
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, People's Republic of China.
| |
Collapse
|
44
|
Lama-Sherpa TD, Lin VTG, Metge BJ, Weeks SE, Chen D, Samant RS, Shevde LA. Hedgehog signaling enables repair of ribosomal DNA double-strand breaks. Nucleic Acids Res 2020; 48:10342-10352. [PMID: 32894284 PMCID: PMC7544215 DOI: 10.1093/nar/gkaa733] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/20/2020] [Accepted: 09/04/2020] [Indexed: 12/22/2022] Open
Abstract
Ribosomal DNA (rDNA) consists of highly repeated sequences that are prone to incurring damage. Delays or failure of rDNA double-strand break (DSB) repair are deleterious, and can lead to rDNA transcriptional arrest, chromosomal translocations, genomic losses, and cell death. Here, we show that the zinc-finger transcription factor GLI1, a terminal effector of the Hedgehog (Hh) pathway, is required for the repair of rDNA DSBs. We found that GLI1 is activated in triple-negative breast cancer cells in response to ionizing radiation (IR) and localizes to rDNA sequences in response to both global DSBs generated by IR and site-specific DSBs in rDNA. Inhibiting GLI1 interferes with rDNA DSB repair and impacts RNA polymerase I activity and cell viability. Our findings tie Hh signaling to rDNA repair and this heretofore unknown function may be critically important in proliferating cancer cells.
Collapse
Affiliation(s)
| | - Victor T G Lin
- Division of Hematology and Oncology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brandon J Metge
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shannon E Weeks
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dongquan Chen
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Preventative Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rajeev S Samant
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
- Birmingham VA Medical Center, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
45
|
Brueffer C, Gladchuk S, Winter C, Vallon‐Christersson J, Hegardt C, Häkkinen J, George AM, Chen Y, Ehinger A, Larsson C, Loman N, Malmberg M, Rydén L, Borg Å, Saal LH. The mutational landscape of the SCAN-B real-world primary breast cancer transcriptome. EMBO Mol Med 2020; 12:e12118. [PMID: 32926574 PMCID: PMC7539222 DOI: 10.15252/emmm.202012118] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 08/08/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is a disease of genomic alterations, of which the panorama of somatic mutations and how these relate to subtypes and therapy response is incompletely understood. Within SCAN-B (ClinicalTrials.gov: NCT02306096), a prospective study elucidating the transcriptomic profiles for thousands of breast cancers, we developed a RNA-seq pipeline for detection of SNVs/indels and profiled a real-world cohort of 3,217 breast tumors. We describe the mutational landscape of primary breast cancer viewed through the transcriptome of a large population-based cohort and relate it to patient survival. We demonstrate that RNA-seq can be used to call mutations in genes such as PIK3CA, TP53, and ERBB2, as well as the status of molecular pathways and mutational burden, and identify potentially druggable mutations in 86.8% of tumors. To make this rich dataset available for the research community, we developed an open source web application, the SCAN-B MutationExplorer (http://oncogenomics.bmc.lu.se/MutationExplorer). These results add another dimension to the use of RNA-seq as a clinical tool, where both gene expression- and mutation-based biomarkers can be interrogated in real-time within 1 week of tumor sampling.
Collapse
Affiliation(s)
- Christian Brueffer
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
| | - Sergii Gladchuk
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
| | - Christof Winter
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
- Present address:
Institut für Klinische Chemie und PathobiochemieKlinikum rechts der IsarTechnische Universität MünchenMünchenGermany
| | - Johan Vallon‐Christersson
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
- CREATE Health Strategic Center for Translational Cancer ResearchLund UniversityLundSweden
| | - Cecilia Hegardt
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
- CREATE Health Strategic Center for Translational Cancer ResearchLund UniversityLundSweden
| | - Jari Häkkinen
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
| | - Anthony M George
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
| | - Yilun Chen
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
| | - Anna Ehinger
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
- Department of PathologySkåne University HospitalLundSweden
| | - Christer Larsson
- Lund University Cancer CenterLundSweden
- Division of Molecular PathologyDepartment of Laboratory MedicineLund UniversityLundSweden
| | - Niklas Loman
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
- Department of OncologySkåne University HospitalLundSweden
| | | | - Lisa Rydén
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
- Department of SurgerySkåne University HospitalLundSweden
| | - Åke Borg
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
- CREATE Health Strategic Center for Translational Cancer ResearchLund UniversityLundSweden
| | - Lao H Saal
- Division of OncologyDepartment of Clinical SciencesLund UniversityLundSweden
- Lund University Cancer CenterLundSweden
- CREATE Health Strategic Center for Translational Cancer ResearchLund UniversityLundSweden
| |
Collapse
|
46
|
Guimaraes VSN, Vidal MTA, de Faro Valverde L, de Oliveira MG, de Oliveira Siquara da Rocha L, Coelho PLC, Soares FA, de Freitas Souza BS, Bezerra DP, Coletta RD, Pereira TA, Dos Santos JN, Gurgel Rocha CA. Hedgehog pathway activation in oral squamous cell carcinoma: cancer-associated fibroblasts exhibit nuclear GLI-1 localization. J Mol Histol 2020; 51:675-684. [PMID: 33000351 DOI: 10.1007/s10735-020-09913-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/22/2020] [Indexed: 01/09/2023]
Abstract
The purpose of this study was to evaluate the expression of Hedgehog (HH) signaling molecules (SHH and GLI-1) by cancer-associated fibroblasts (CAF) in oral squamous cell carcinoma (OSCC). Immunohistochemistry was used to detect molecular HH signaling and CAF-related protein expression, including α-SMA and S100A4, in 70 samples of human OSCC. The colocalization of α-SMA and S100A4 with SHH was also evaluated by double-staining. In vitro study was performed using primary normal oral fibroblast (NOF) and CAF through immunofluorescence and Western Blot for CAF-proteins, SHH, and GLI-1. Forty-five cases (64.28%) were positive for α-SMA exclusively in tumor stroma, and S100A4 was identified in the cytoplasm of CAFs in 94.28% (n = 66) of the cases. With respect to stromal cells, 64 (91.43%) OSCC cases were positive for SHH, and 31 were positive for GLI-1 (44.29%); positive correlations were found between SHH and α-SMA (p < 0.0001, φ = 0.51), as well as between SHH and S100A4 (p = 0.087, φ = 0.94). Protein expression of SHH and GLI-1 was observed in primary CAFs and NOFs. Although SHH was found to be localized in the cellular cytoplasm of both cell types, GLI-1 was present only in the nuclei of CAF. Our results indicate that CAFs are not only potential sources of HH ligands in tumor stroma, but may also respond to HH signaling through nuclear GLI-1 activation. We further observed that elevated SHH expression by OSCC cells was associated with higher CAF density, reinforcing the chemoattractant role played by these molecules.
Collapse
Affiliation(s)
- Vanessa Sousa Nazare Guimaraes
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Waldemar Falcao Street, 121, Candeal, Salvador, Bahia, 40296-710, Brazil
- Federal University of Bahia, Augusto Viana Street, s/n, Canela, Salvador, Bahia, 40110-909, Brazil
| | - Manuela Torres Andion Vidal
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Waldemar Falcao Street, 121, Candeal, Salvador, Bahia, 40296-710, Brazil
- Federal University of Bahia, Augusto Viana Street, s/n, Canela, Salvador, Bahia, 40110-909, Brazil
| | - Ludmila de Faro Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Waldemar Falcao Street, 121, Candeal, Salvador, Bahia, 40296-710, Brazil
- Federal University of Bahia, Augusto Viana Street, s/n, Canela, Salvador, Bahia, 40110-909, Brazil
| | - Marbele Guimarães de Oliveira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Waldemar Falcao Street, 121, Candeal, Salvador, Bahia, 40296-710, Brazil
- Federal University of Bahia, Augusto Viana Street, s/n, Canela, Salvador, Bahia, 40110-909, Brazil
| | - Leonardo de Oliveira Siquara da Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Waldemar Falcao Street, 121, Candeal, Salvador, Bahia, 40296-710, Brazil
- Federal University of Bahia, Augusto Viana Street, s/n, Canela, Salvador, Bahia, 40110-909, Brazil
| | - Paulo Lucas Cerqueira Coelho
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Waldemar Falcao Street, 121, Candeal, Salvador, Bahia, 40296-710, Brazil
| | - Fernando Augusto Soares
- D'Or Institute for Research and Education, Diniz Cordeiro Street, 30, Botafogo, Rio de Janeiro, 22281-100, Brazil
- School of Dentistry, University of São Paulo, Prof. Almeida Prado Avenue, 1280, Butantã, São Paulo, 05508-900, Brazil
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Waldemar Falcao Street, 121, Candeal, Salvador, Bahia, 40296-710, Brazil
| | - Daniel Pereira Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Waldemar Falcao Street, 121, Candeal, Salvador, Bahia, 40296-710, Brazil
| | - Ricardo D Coletta
- School of Dentistry, University of Campinas, Limeira Avenue, 901, Areiao, Piracicaba, São Paulo, 13414-903, Brazil
| | - Thiago Almeida Pereira
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305-5463, USA
| | - Jean Nunes Dos Santos
- Federal University of Bahia, Augusto Viana Street, s/n, Canela, Salvador, Bahia, 40110-909, Brazil
| | - Clarissa Araújo Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Waldemar Falcao Street, 121, Candeal, Salvador, Bahia, 40296-710, Brazil.
- Federal University of Bahia, Augusto Viana Street, s/n, Canela, Salvador, Bahia, 40110-909, Brazil.
- Laboratory of Pathology and Molecular Biology, Gonçalo Moniz Institute, Oswaldo Cruz Foundation,121 Waldemar Falcao, Candeal, Salvador, Bahia, Brazil.
| |
Collapse
|
47
|
Samuel SM, Varghese E, Koklesová L, Líšková A, Kubatka P, Büsselberg D. Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells. Cancers (Basel) 2020; 12:E2482. [PMID: 32883003 PMCID: PMC7565921 DOI: 10.3390/cancers12092482] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the leaps and bounds in achieving success in the management and treatment of breast cancers through surgery, chemotherapy, and radiotherapy, breast cancer remains the most frequently occurring cancer in women and the most common cause of cancer-related deaths among women. Systemic therapeutic approaches, such as chemotherapy, although beneficial in treating and curing breast cancer subjects with localized breast tumors, tend to fail in metastatic cases of the disease due to (a) an acquired resistance to the chemotherapeutic drug and (b) the development of intrinsic resistance to therapy. The existence of cancer stem cells (CSCs) plays a crucial role in both acquired and intrinsic chemoresistance. CSCs are less abundant than terminally differentiated cancer cells and confer chemoresistance through a unique altered metabolism and capability to evade the immune response system. Furthermore, CSCs possess active DNA repair systems, transporters that support multidrug resistance (MDR), advanced detoxification processes, and the ability to self-renew and differentiate into tumor progenitor cells, thereby supporting cancer invasion, metastasis, and recurrence/relapse. Hence, current research is focusing on targeting CSCs to overcome resistance and improve the efficacy of the treatment and management of breast cancer. Studies revealed that metformin (1, 1-dimethylbiguanide), a widely used anti-hyperglycemic agent, sensitizes tumor response to various chemotherapeutic drugs. Metformin selectively targets CSCs and improves the hypoxic microenvironment, suppresses the tumor metastasis and inflammation, as well as regulates the metabolic programming, induces apoptosis, and reverses epithelial-mesenchymal transition and MDR. Here, we discuss cancer (breast cancer) and chemoresistance, the molecular mechanisms of chemoresistance in breast cancers, and metformin as a chemo-sensitizing/re-sensitizing agent, with a particular focus on breast CSCs as a critical contributing factor to acquired and intrinsic chemoresistance. The review outlines the prospects and directions for a better understanding and re-purposing of metformin as an anti-cancer/chemo-sensitizing drug in the treatment of breast cancer. It intends to provide a rationale for the use of metformin as a combinatory therapy in a clinical setting.
Collapse
Affiliation(s)
- Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Lenka Koklesová
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.)
| | - Alena Líšková
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| |
Collapse
|
48
|
Oliphant MUJ, Kong D, Zhou H, Lewis MT, Ford HL. Two Sides of the Same Coin: The Role of Developmental pathways and pluripotency factors in normal mammary stem cells and breast cancer metastasis. J Mammary Gland Biol Neoplasia 2020; 25:85-102. [PMID: 32323111 PMCID: PMC7395869 DOI: 10.1007/s10911-020-09449-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer initiation and progression are often observed as the result of dysregulation of normal developmental processes and pathways. Studies focused on normal mammary stem/progenitor cell activity have led to an understanding of how breast cancer cells acquire stemness-associated properties including tumor initiation, survival and multi-lineage differentiation into heterogeneous tumors that become difficult to target therapeutically. Importantly, more recent investigations have provided valuable insight into how key developmental regulators can impact multiple phases of metastasis, where they are repurposed to not only promote metastatic phenotypes such as migration, invasion and EMT at the primary site, but also to regulate the survival, initiation and maintenance of metastatic lesions at secondary organs. Herein, we discuss findings that have led to a better understanding of how embryonic and pluripotency factors contribute not only to normal mammary development, but also to metastatic progression. We further examine the therapeutic potential of targeting these developmental pathways, and discuss how a better understanding of compensatory mechanisms, crosstalk between pathways, and novel experimental models could provide critical insight into how we might exploit embryonic and pluripotency regulators to inhibit tumor progression and metastasis.
Collapse
Affiliation(s)
- M U J Oliphant
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Department of Cell Biology and Ludwig Center at Harvard, Harvard Medical School, 240 Longwood Avenue, Building C1, Room 513B, Boston, MA, 02115, USA
| | - Deguang Kong
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
| | - Hengbo Zhou
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
| | - M T Lewis
- Departments of Molecular and Cellular Biology and Radiology. Lester and Sue Smith Breast Center, Baylor College of Medicine. One Baylor Plaza BCM600, Room N1210, Houston, TX, 77030, USA
| | - H L Ford
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
| |
Collapse
|
49
|
Singh AN, Sharma N. Epigenetic Modulators as Potential Multi-targeted Drugs Against Hedgehog Pathway for Treatment of Cancer. Protein J 2020; 38:537-550. [PMID: 30993446 DOI: 10.1007/s10930-019-09832-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Sonic hedgehog signalling is known to play a crucial role in regulating embryonic development, cancer stem cell maintenance and tissue patterning. Dysregulated hedgehog signalling has been reported to affect tumorigenesis and drug response in various human malignancies. Epigenetic therapy relying on DNA methyltransferase and Histone deacetylase inhibitors are being proposed as potential drug candidates considering their efficiency in preventing development of cancer progenitor cells, killing drug resistant cells and also dictating "on/off" switch of tumor suppressor genes and oncogenes. In this docking approach, epigenetic modulators were virtually screened for their efficiency in inhibiting key regulators of SHH pathway viz., sonic hedgehog, Smoothened and Gli using polypharmacological approach. The control drugs and epigenetic modulators were docked with PDB protein structures using AutoDock vina and further checked for their drug-likeness properties. Further molecular dynamics simulation using VMD and NAMD, and MMP/GBSA energy calculation were employed for verifying the stability and entropy of the ligand-receptor complex. EPZ-6438 and GSK 343 (EZH2 inhibitors), CHR 3996 and Mocetinostat (HDAC inhibitors), GSK 126 (HKMT inhibitor) and UNC 1215 (L3MBTL3 antagonist) exhibited multiple-targeted approach in modulating HH signalling. This is the first study to report these epigenetic drugs as potential multi-targeted hedgehog pathway inhibitors. Thus, epigenetic polypharmacology approach can be explored as a better alternative to challenges of acute long term toxicity and drug resistance occurring due to traditional single targeted chemotherapy in the future.
Collapse
Affiliation(s)
- Anshika N Singh
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Gram-Lavale, Taluka-Mulshi, Pune, 412115, India
| | - Neeti Sharma
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Gram-Lavale, Taluka-Mulshi, Pune, 412115, India.
| |
Collapse
|
50
|
Malla RR, Deepak K, Merchant N, Dasari VR. Breast Tumor Microenvironment: Emerging target of therapeutic phytochemicals. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 70:153227. [PMID: 32339885 DOI: 10.1016/j.phymed.2020.153227] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/31/2020] [Accepted: 04/11/2020] [Indexed: 06/11/2023]
Abstract
Triple negative breast cancer (TNBC) is the most aggressive and challenging form of breast cancers. Tumor microenvironment (TME) of TNBC is associated with induction of metastasis, immune system suppression, escaping immune detection and drug resistance. TME is highly complex and heterogeneous, consists of tumor cells, stromal cells and immune cells. The rapid expansion of tumors induce hypoxia, which concerns the reprogramming of TME components. The reciprocal communication of tumor cells and TME cells predisposes cancer cells to metastasis by modulation of developmental pathways, Wnt, notch, hedgehog and their related mechanisms in TME. Dietary phytochemicals are non-toxic and associated with various human health benefits and remarkable spectrum of biological activities. The phytochemicals serve as vital resources for drug discovery and also as a source for breast cancer therapy. The novel properties of dietary phytochemicals propose platform for modulation of tumor signaling, overcoming drug resistance, and targeting TME. Therefore, TME could serve as promising target for the treatment of TNBC. This review presents current status and implications of experimentally evaluated therapeutic phytochemicals as potential targeting agents of TME, potential nanosystems for targeted delivery of phytochemicals and their current challenges and future implications in TNBC treatment. The dietary phytochemicals especially curcumin with significant delivery system could prevent TNBC development as it is considered safe and well tolerated in phase II clinical trials.
Collapse
Affiliation(s)
- Rama Rao Malla
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, India.
| | - Kgk Deepak
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, India
| | - Neha Merchant
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Venkata Ramesh Dasari
- Department of Molecular and Functional Genomics, Geisinger Clinic, 100 Academy Ave, Danville, PA, 17822, USA
| |
Collapse
|