1
|
Chang TM, Fang WY, Hsu HP, Chu PY, Jiang SS, Huang KW, Hung WC, Lin HY, Tsai HJ. PCK2 promotes invasion and epithelial-to-mesenchymal transition in triple-negative breast cancer by promoting TGF-β/SMAD3 signaling through inhibiting TRIM67-mediated SMAD3 ubiquitination. Cancer Biol Ther 2025; 26:2478670. [PMID: 40081967 PMCID: PMC11913380 DOI: 10.1080/15384047.2025.2478670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/22/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
PCK2, which encodes mitochondrial phosphoenolpyruvate carboxykinase (PEPCK-M), is upregulated in various cancers. We demonstrated high expression of PEPCK-M in approximately half of triple-negative breast cancers (TNBCs) previously. TNBC is associated with an aggressive phenotype and a high metastasis rate. In this study, we investigated the role of PCK2 in TNBC. PCK2 knockdown suppressed proliferation and mTOR signaling in TNBC cells. In addition, cell invasion/migration ability and the expression of epithelial-to-mesenchymal transition (EMT) markers were positively correlated with PCK2 expression in TNBC cells via regulation of transforming growth factor-β (TGF-β)/SMAD3 signaling. SMAD3 was positively regulated by PCK2 in TNBC cells. Knockdown of SMAD3 in PCK2-overexpressing TNBC cells reduced the expression levels of EMT markers, Snail and Slug, and suppressed cell invasion/migration. In addition, PCK2 knockdown attenuated the stimulatory effect of TGF-β on SMAD3 phosphorylation in TNBC cells. PEPCK-M promotes the protein and mRNA expression of SMAD3 via competitive binding to tripartite motif-containing 67 (TRIM67), an E3 ubiquitin ligase, to reduce SMAD3 ubiquitination, which leads to promoting nuclear translocation of SMAD3 and autoregulation of SMAD3 transcription. Moreover, high PCK2 mRNA expression was significantly associated with poor survival in TNBC patients. In conclusion, our study revealed for the first time that PCK2 activates TGF-β/SMAD3 signaling by regulating the expression and phosphorylation of SMAD3 by inhibiting TRIM67-mediated SMAD3 ubiquitination and promoting the stimulatory effect of TGF-β to promote TNBC invasion. The regulatory effect of PCK2 on mTOR and TGF-β/SMAD3 signaling suggests that PCK2 is a potential therapeutic target for suppressing TNBC progression.
Collapse
Affiliation(s)
- Tsung-Ming Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung, Taiwan
| | - Wei-Yu Fang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Pei-Yi Chu
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Shih Sheng Jiang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Kuo-Wei Huang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Hui-You Lin
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Hui-Jen Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
2
|
Ha J. DeepWalk-Based Graph Embeddings for miRNA-Disease Association Prediction Using Deep Neural Network. Biomedicines 2025; 13:536. [PMID: 40149513 PMCID: PMC11940379 DOI: 10.3390/biomedicines13030536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
Background: In recent years, micro ribonucleic acids (miRNAs) have been recognized as key regulators in numerous biological processes, particularly in the development and progression of diseases. As a result, extensive research has focused on uncovering the critical involvement of miRNAs in disease mechanisms to better comprehend the underlying causes of human diseases. Despite these efforts, relying solely on biological experiments to identify miRNA-disease associations is both time-consuming and costly, making it an impractical approach for large-scale studies. Methods: In this paper, we propose a novel DeepWalk-based graph embedding method for predicting miRNA-disease association (DWMDA). Using DeepWalk, we extracted meaningful low-dimensional vectors from the miRNA and disease networks. Then, we applied a deep neural network to identify miRNA-disease associations using the low-dimensional vectors of miRNAs and diseases extracted via DeepWalk. Results: An ablation study was conducted to assess the proposed graph embedding modules. Furthermore, DWMDA demonstrates exceptional performance in two major cancer case studies (breast and lung), with results based on statistically robust measures, further emphasizing its reliability as a method for identifying associations between miRNAs and diseases. Conclusions: We expect that our model will not only facilitate the accurate prediction of disease-associated miRNAs but also serve as a generalizable framework for exploring interactions among various biological entities.
Collapse
Affiliation(s)
- Jihwan Ha
- Major of Big Data Convergence, Division of Data Information Science, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
3
|
Ghosh P, Dey A, Nandi S, Majumder R, Das S, Mandal M. CTGF (CCN2): a multifaceted mediator in breast cancer progression and therapeutic targeting. Cancer Metastasis Rev 2025; 44:32. [PMID: 39945880 DOI: 10.1007/s10555-025-10248-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 02/01/2025] [Indexed: 03/28/2025]
Abstract
Breast cancer, with its diverse subtypes like ER-positive, HER-2-positive, and triple-negative, presents complex challenges demanding personalized treatment approaches. The intricate interplay of genetic, environmental, and lifestyle factors underscores its status as a primary contributor to cancer-related fatalities in women globally. Understanding the molecular drivers specific to each subtype is crucial for developing effective therapies. In this landscape, connective tissue growth factor (CTGF), also referred to as cellular communication network factor 2 (CCN2), emerges as a significant player. CTGF regulates critical biological activities like cell growth, invasion, and migration, impacting breast cancer development and progression. It modulates breast tumor microenvironment by promoting angiogenesis, activating cancer-associated fibroblasts (CAFs), and inducing inflammation. The activity of CTGF depends on several factors including oxygen levels, hormone signals, and growth factors and differs according to the type of breast cancer. CTGF can regulate breast cancer cells by activating various signaling pathways and modulating the transcription of other genes that are involved in tumor development and metastasis including S100A4, glucose transporter 3 (GLUT3), and vascular endothelial growth factor (VEGF). The matricellular protein can be considered a potential therapeutic target, as it can promote tumor growth and confer drug resistance in breast cancer. Numerous tactics, including neutralizing antibodies, antisense oligonucleotides, natural compounds, recombinant proteins, and short hairpin RNAs have been suggested to block its function. This review highlights the structure of CTGF, regulation of its expression, and current knowledge of its oncogenic role in breast cancer, as well as focusing on potential therapeutic strategies for targeting CTGF in breast cancer.
Collapse
Affiliation(s)
- Priya Ghosh
- Cancer Biology Lab, School of Medical Science & Technology, Indian Institute of Technology, Kharagpur 721302, Kharagpur, West Bengal, India
| | - Ankita Dey
- Cancer Biology Lab, School of Medical Science & Technology, Indian Institute of Technology, Kharagpur 721302, Kharagpur, West Bengal, India
| | - Suvendu Nandi
- Cancer Biology Lab, School of Medical Science & Technology, Indian Institute of Technology, Kharagpur 721302, Kharagpur, West Bengal, India
| | - Ranabir Majumder
- Cancer Biology Lab, School of Medical Science & Technology, Indian Institute of Technology, Kharagpur 721302, Kharagpur, West Bengal, India
| | - Subhayan Das
- Department of Allied Health Sciences, Brainware University, Kolkata 700125, Barasat, West Bengal, India
| | - Mahitosh Mandal
- Cancer Biology Lab, School of Medical Science & Technology, Indian Institute of Technology, Kharagpur 721302, Kharagpur, West Bengal, India.
| |
Collapse
|
4
|
Al Shahrani M, Abohassan M, Alshahrani M, Gahtani RM, Rajagopalan P. Identification of 8-(2-methyl phenyl)-9H-benzo[f]indeno[2,1-c]quinolin-9-one (C-5635020) as a novel and selective TGFβ RII kinase inhibitor for breast cancer therapy. Biochem Biophys Res Commun 2025; 746:151225. [PMID: 39761620 DOI: 10.1016/j.bbrc.2024.151225] [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: 08/24/2024] [Revised: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 01/15/2025]
Abstract
OBJECTIVE AND SIGNIFICANCE Transforming growth factor-beta (TGF-β) plays a pivotal role in breast development by modulating tissue composition during the developmental phase. The TGFβ type II receptor (TGFβ RII) is implicated in breast cancer and represents a valuable therapeutic target. Due to the off-target side effects of many existing TGFβI/TGFβ RII inhibitors, a more targeted approach to drug discovery is necessary. This study used computational modeling and molecular dynamics simulations to screen the ChemBridge small molecule library against TGFβ RII. METHODS This study employed high-throughput virtual screening, molecular dynamics simulations, and binding free energy calculations to identify potential inhibitors targeting TGF-β RII. MDA-MB 231 and MCF-7 breast cancer cells were used in anti-proliferative, tans-endothelial migration, and flow cytometric assays for in vitro validations. RESULTS We identified 8-(2-methylphenyl)-9H-benzo[f]indeno[2,1-c]quinolin-9-one (C-5635020) as a potent and selective inhibitor. Protein-ligand modeling analysis revealed that C-5635020 targets the kinase domain of TGFβ RII with superior binding affinities compared to the standard drug, staurosporine. Computational results suggest that C-5635020 selectively binds and inhibits TGFβ RII activity, thereby controlling cell proliferation in breast cancer. In vitro, experiments corroborated these predictions, where C-5635020 inhibited TGFβ RII and p-Smad 2/3 positive population in MDAMB-231 and MCF-7 cells. The compound dose-dependently inhibited cell proliferation, trans-endothelial migration, and increased apoptosis in both breast cancer cell lines. CONCLUSION The strong binding affinity, stability, and favorable thermodynamics of C-5635020 with established in vitro efficacy highlight its potential as a lead compound for further preclinical and clinical developments for breast cancer treatment.
Collapse
Affiliation(s)
- Mesfer Al Shahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Reem M Gahtani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Prasanna Rajagopalan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| |
Collapse
|
5
|
Mariana SM, Brenda RP, Heriberto PG, Cristina L, David B, Guadalupe ÁL. GPER1 activation by estrogenic compounds in the inflammatory profile of breast cancer cells. J Steroid Biochem Mol Biol 2025; 245:106639. [PMID: 39571822 DOI: 10.1016/j.jsbmb.2024.106639] [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: 08/08/2024] [Revised: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Breast cancer (BC) is the most frequent female neoplasm worldwide. Its establishment and development have been related to inflammatory cytokine expression. Steroid hormones such as estradiol (E2) can regulate proinflammatory cytokine secretion through interaction with its nuclear receptors. However, little is known regarding the activation of its membrane estrogen receptor (GPER1) and the inflammatory cytokine environment in BC. We have studied the synthesis and biological effects of molecules analogs to E2 for hormone replacement therapy (HRT), such as pentolame. Nevertheless, its interaction with GPER1 and the modulation of inflammatory cytokines in different BC types has been barely studied and deserves deeper investigation. In this research, the role of GPER1 in the proliferation and modulation of inflammatory cytokines involved in carcinogenesis and metastatic processes in different BC cell lines was assessed by binding to various compounds. To achieve this goal, the presence of GPER1 was identified in different BC cell lines. Subsequently, cell proliferation after exposure to E2, pentolame and GPER1 agonist, G1, was subsequently determined alone or in combination with the GPER1 antagonist, G15. Finally, the pro-inflammatory cytokine secretion derived from the supernatants of BC cells exposed to the previous treatments was also assessed. Interestingly, GPER1 activation or inhibition has significant effects on the cytokine regulation associated with invasion in BC. Notably, pentolame did not induce cell proliferation or increase the proinflammatory cytokine expression compared to E2 in BC cell lines. In addition, pentolame did not induce the presence of the cell adhesion molecule PECAM-1. In contrast, E2 treatment weakly induced the expression of PECAM-1 in MCF-7 and HCC1937 cells, and G1 treatment showed this effect only in MCF-7 cells. The results suggest that GPER1 might be a significant inflammatory modulator with angiogenic-related effects in BC cells. In addition, pentolame might represent an HRT alternative in patients with BC predisposition.
Collapse
Affiliation(s)
- Segovia-Mendoza Mariana
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - Reyes-Plata Brenda
- Facultad de Estudios Superiores Zaragoza. Universidad Nacional Autónoma de México,Ciudad de México, Mexico
| | - Prado-Garcia Heriberto
- Laboratorio de Onco-Inmunobiología, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias, "Ismael Cosio Villegas" Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México 14080, Mexico
| | - Lemini Cristina
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Barrera David
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Col. Belisario Domínguez, Sección XVI, Ciudad de México 14080, Mexico
| | - Ángeles-López Guadalupe
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
6
|
Gehling GM, Alfaqih M, Pruinelli L, Starkweather A, Dungan JR. A systematic review of candidate genes and their relevant pathways for metastasis among adults diagnosed with breast cancer. Breast Cancer Res 2024; 26:165. [PMID: 39593069 PMCID: PMC11590482 DOI: 10.1186/s13058-024-01914-6] [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: 08/24/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Presently incurable, metastatic breast cancer is estimated to occur in as many as 30% of those diagnosed with early-stage breast cancer. Timely and accurate identification of those at risk for developing metastasis using validated biomarkers has the potential to have profound impact on overall survival rates. Our primary goal was to conduct a systematic review and synthesize the existing body of scientific knowledge on the candidate genes and their respective single nucleotide polymorphisms associated with metastasis-related outcomes among patients diagnosed with breast cancer. This knowledge is critical to inform future hypothesis-driven and validation research aimed at enhancing clinical decision-making for breast cancer patients. METHODS Using PRISMA guidelines, literature searches were conducted on September 13th, 2023, using PubMed and Embase databases. The systematic review protocol was registered with INPLASY (DOI: https://doi.org/10.37766/inplasy2024.8.0014 ). Covidence software was used to facilitate the screening and article extraction processes. Peer-reviewed articles were selected if authors reported on single nucleotide polymorphisms directly associated with metastasis among adults diagnosed with breast cancer. FINDINGS We identified 451 articles after 44 duplicates were removed resulting in 407 articles to be screened for study inclusion. Three reviewers completed the article screening process which resulted in 86 articles meeting the study inclusion criteria. Sampling varied across studies with the majority utilizing a case-control design (n = 75, 87.2%), with sample sizes ranging from 23 to 1,017 participants having mean age 50.65 ± 4.50 (min-max: 20-75). The synthesis of this internationally generated evidence revealed that the scientific area on the underlying biological contributions to breast cancer metastasis remains predominantly exploratory in nature (n = 74, 86%). Of the 12 studies with reported power analyses, only 9 explicitly stated the power values which ranged from 47.88 to 99%. DISCUSSION Understanding the underlying biological mechanisms contributing to metastasis is a critical component for precision oncological therapeutics and treatment approaches. Current evidence investigating the contribution of SNPs to the development of metastasis is characterized by underpowered candidate gene studies. To inform individualized precision health practices and improve breast cancer survival outcomes, future hypothesis-driven research is needed to replicate these associations in larger, more diverse datasets.
Collapse
Affiliation(s)
- Gina M Gehling
- College of Nursing, University of Florida, 1225 Center Dr, PO BOX 100197, Gainesville, FL, 32610-1097, USA
| | - Miad Alfaqih
- College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lisiane Pruinelli
- College of Nursing, University of Florida, 1225 Center Dr, PO BOX 100197, Gainesville, FL, 32610-1097, USA
| | - Angela Starkweather
- College of Nursing, University of Florida, 1225 Center Dr, PO BOX 100197, Gainesville, FL, 32610-1097, USA
| | - Jennifer R Dungan
- College of Nursing, University of Florida, 1225 Center Dr, PO BOX 100197, Gainesville, FL, 32610-1097, USA.
| |
Collapse
|
7
|
Bong AHL, Robitaille M, Lin S, McCart-Reed A, Milevskiy M, Angers S, Roberts-Thomson SJ, Monteith GR. TMCO1 is upregulated in breast cancer and regulates the response to pro-apoptotic agents in breast cancer cells. Cell Death Discov 2024; 10:421. [PMID: 39353922 PMCID: PMC11445413 DOI: 10.1038/s41420-024-02183-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
The release of Ca2+ ions from endoplasmic reticulum calcium stores is a key event in a variety of cellular processes, including gene transcription, migration and proliferation. This release of Ca2+ often occurs through inositol 1,4,5-triphosphate receptors and the activity of these channels and the levels of stored Ca2+ in the endoplasmic reticulum are important regulators of cell death in cancer cells. A recently identified Ca2+ channel of the endoplasmic reticulum is transmembrane and coiled-coil domains 1 (TMCO1). In this study, we link the overexpression of TMCO1 with prognosis in node-positive basal breast cancer patients. We also identify interacting proteins of TMCO1, which include endoplasmic reticulum-resident proteins involved in Ca2+ regulation and proteins directly involved in nucleocytoplasmic transport. Interacting proteins included nuclear transport proteins and TMCO1 was shown to have both nuclear and endoplasmic reticulum localisation in MDA-MB-231 basal breast cancer cells. These studies also define a role for TMCO1 in the regulation of breast cancer cells in their sensitivity to BCL-2/MCL-1 inhibitors, analogous to the role of inositol 1,4,5-triphosphate receptors in the regulation of cell death pathways activated by these agents.
Collapse
Affiliation(s)
- Alice H L Bong
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Mélanie Robitaille
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Sichun Lin
- Donelly Centre, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Amy McCart-Reed
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Michael Milevskiy
- ACRF Cancer Biology and Stem Cells, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Stéphane Angers
- Donelly Centre, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 1A2, Canada
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | | | - Gregory R Monteith
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
| |
Collapse
|
8
|
Zhang X, Miao J, Song Y, Zhang J, Miao M. Review on effects and mechanisms of plant-derived natural products against breast cancer bone metastasis. Heliyon 2024; 10:e37894. [PMID: 39318810 PMCID: PMC11420494 DOI: 10.1016/j.heliyon.2024.e37894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/16/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Bone metastasis is the prevalent form of metastasis in breast cancer, resulting in severe pain, pathological fractures, nerve compression, hypercalcemia, and other complications that significantly impair patients' quality of life. The infiltration and colonization of breast cancer (BC) cells in bone tissue disrupt the delicate balance between osteoblasts and osteoclasts within the bone microenvironment, initiating a vicious cycle of bone metastasis. Once bone metastasis occurs, conventional medical therapy with bone-modifying agents is commonly used to alleviate bone-related complications and improve patients' quality of life. However, the utilization of bone-modifying agents may cause severe drug-related adverse effects. Plant-derived natural products such as terpenoids, alkaloids, coumarins, and phenols have anti-tumor, anti-inflammatory, and anti-angiogenic pharmacological properties with minimal side effects. Certain natural products that exhibit both anti-breast cancer and anti-bone metastasis effects are potential therapeutic agents for breast cancer bone metastasis (BCBM). This article reviewed the effects of plant-derived natural products against BCBM and their mechanisms to provide a reference for the research and development of drugs related to BCBM.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jinxin Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yagang Song
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jiawen Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| |
Collapse
|
9
|
Kochumon S, Al-Sayyar A, Jacob T, Bahman F, Akhter N, Wilson A, Sindhu S, Hannun YA, Ahmad R, Al-Mulla F. TGF-β and TNF-α interaction promotes the expression of MMP-9 through H3K36 dimethylation: implications in breast cancer metastasis. Front Immunol 2024; 15:1430187. [PMID: 39351229 PMCID: PMC11439675 DOI: 10.3389/fimmu.2024.1430187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/26/2024] [Indexed: 10/04/2024] Open
Abstract
Increased MMP-9 expression in the tumor microenvironment (TME) plays a crucial role in the extracellular matrix remodeling to facilitate cancer invasion and metastasis. However, the mechanism of MMP-9 upregulation in TME remains elusive. Since TGF-β and TNF-α levels are elevated in TME, we asked whether these two agents interacted to induce/augment MMP-9 expression. Using a well-established MDA-MB-231 breast cancer model, we found that the synergy between TGF-β and TNF-α led to MMP-9 upregulation at the transcriptional and translational levels, compared to treatments with each agent alone. Our in vitro findings are corroborated by co-expression of elevated MMP-9 with TGF-β and TNF-α in human breast cancer tissues. Mechanistically, we found that the MMP-9 upregulation driven by TGF-β/TNF-α cooperativity was attenuated by selective inhibition of the TGF-βRI/Smad3 pathway. Comparable outcomes were observed upon inhibition of TGF-β-induced phosphorylation of Smad2/3 and p38. As expected, the cells defective in Smad2/3 or p38-mediated signaling did not exhibit this synergistic induction of MMP-9. Importantly, the inhibition of histone methylation but not acetylation dampened the synergistic MMP-9 expression. Histone modification profiling further identified the H3K36me2 as an epigenetic regulatory mark of this synergy. Moreover, TGF-β/TNF-α co-stimulation led to increased levels of the transcriptionally permissive dimethylation mark at H3K36 in the MMP-9 promoter. Comparable outcomes were noted in cells deficient in NSD2 histone methyltransferase. In conclusion, our findings support a cooperativity model in which TGF-β could amplify the TNF-α-mediated MMP-9 production via chromatin remodeling and facilitate breast cancer invasion and metastasis.
Collapse
Affiliation(s)
- Shihab Kochumon
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Amnah Al-Sayyar
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, Marseille, France
| | - Texy Jacob
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Fatemah Bahman
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Nadeem Akhter
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Ajit Wilson
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Sardar Sindhu
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Yusuf A. Hannun
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, United States
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Fahd Al-Mulla
- Translational Research, Dasman Diabetes Institute, Dasman, Kuwait
| |
Collapse
|
10
|
Zhao S, Song C, Chen F, Li M. LncRNA XIST/miR-455-3p/HOXC4 axis promotes breast cancer development by activating TGF-β/SMAD signaling pathway. Funct Integr Genomics 2024; 24:159. [PMID: 39261346 DOI: 10.1007/s10142-024-01442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/16/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
Breast cancer is the second primary cause of cancer death among women. Long non-coding RNA (lncRNA) X-inactive specific transcript (XIST) is a central regulator for X chromosome inactivation, and its abnormal expression is a primary feature of breast cancer. So far, the mechanism of XIST in breast cancer has not been fully elucidated. We attempted to illustrate the mechanism of XIST in breast cancer. The expressions of XIST, microRNA-455-3p (miR-455-3p) in breast cancer were measured using quantitative real-time PCR. The expressions of homeobox C4 (HOXC4) were assessed with immunohistochemical and Western blot. Also, the functions of XIST in breast cancer were assessed by Cell Counting Kit-8 analysis, colony formation assay, flow cytometry, Western blot, Transwell, and cell scratch assays. Meanwhile, the mechanism of XIST in breast cancer was validated using database analysis and dual-luciferase reporter assay. Furthermore, the function of XIST in breast cancer in vivo was estimated by tumor xenograft model, immunohistochemical assay, and hematoxylin-eosin staining. XIST and HOXC4 expressions were increased, but miR-455-3p expressions were decreased in breast cancer tissues and cells. Knocking down XIST restrained breast cancer cell proliferation, invasion, migration, epithelial-mesenchymal transformation (EMT), and induced cell cycle arrest at G0/G1. Meanwhile, XIST interacted with miR-455-3p, while miR-455-3p interacted with HOXC4. XIST knockdown repressed breast cancer cell proliferation, invasion, and EMT, while miR-455-3p inhibitor or HOXC4 overexpression abolished those impacts. HOXC4 overexpression also blocked the impacts of miR-455-3p mimic on breast cancer cell malignant behavior. In vivo experimental data further indicated that XIST knockdown repressed breast cancer cell tumorigenic ability, and decreased HOXC4 and p-SMAD3 (TGF-β/SMAD-related protein) expressions.XIST/miR-455-3p/HOXC4 facilitated breast cancer development by activating the TGF-β/SMAD pathway.
Collapse
Affiliation(s)
- Shanshan Zhao
- Department of Oncology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, China
| | - Chen Song
- Department of Oncology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, China
| | - Fengxi Chen
- Department of Oncology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, China
| | - Man Li
- Department of Oncology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, China.
| |
Collapse
|
11
|
Amani MS, Peymani M. Investigating the impact of SMAD2 and SMAD4 downregulation in colorectal cancer and their correlation with immune markers, prognosis, and drug resistance and sensitivity. Mol Biol Rep 2024; 51:831. [PMID: 39037563 DOI: 10.1007/s11033-024-09697-x] [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: 04/11/2024] [Accepted: 06/03/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND While many genes linked to colorectal cancer (CRC) contribute to cancer development, a thorough investigation is needed to explore crucial hub genes yet to be fully studied. A pivotal pathway in CRC is transforming growth factor-beta (TGF-β). This study aimed to assess SMAD2 and SMAD4 gene expression from this pathway. METHODS AND RESULTS Counted data from the Cancer Genome Atlas (TCGA) were examined, comparing 483 tumor and 41 normal samples. Using clinical data, genes impacting overall survival (OS) were evaluated. GSE39582 was employed to confirmed the levels of genes in CRC compared to the normal samples. Additionally, employing unhealthy samples and the RT-qPCR means our outcomes was validated. Finally, PharmacoGx information were utilized to connect the levels of potential genes to drug tolerance and susceptibility. Our findings showed SMAD2 and SMAD4 levels in TGF-β signaling were more significant than other pathway genes. Our findings indicated that the protein levels of these genes were lower in malignant tissues than in healthy tissues. Results revealed a significant correlation between low levels of SMAD2 and unfavorable OS in CRC individuals. RT-qPCR results demonstrated decreased expressions of both SMAD2 and SMAD4 in cancer tissues compared to elevated levels in adjacent normal samples. Our results showed significant association between selected genes and immune cell infiltration markers such as CD8+, and B-cells. Our results indicated a potential association among the levels of SMAD2 and SMAD4 genes and tolerance and susceptibility to Nilotinib and Panobinostat drugs. CONCLUSION Reduced expression of SMAD2 and SMAD4 may be pivotal in CRC progression, impacting downstream genes unrelated to patient OS. These findings suggest a potential role for SMAD2 and SMAD4 as predictive markers for drug response in CRC patients.
Collapse
Affiliation(s)
- Melika Saadat Amani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| |
Collapse
|
12
|
Nong J, Shen S, Hong F, Xiao F, Meng L, Li P, Lei X, Chen YG. Verteporfin inhibits TGF-β signaling by disrupting the Smad2/3-Smad4 interaction. Mol Biol Cell 2024; 35:ar95. [PMID: 38696259 PMCID: PMC11244160 DOI: 10.1091/mbc.e24-02-0073] [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: 02/20/2024] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/04/2024] Open
Abstract
Transforming growth factor-β (TGF-β) signaling plays a crucial role in pathogenesis, such as accelerating tissue fibrosis and promoting tumor development at the later stages of tumorigenesis by promoting epithelial-mesenchymal transition (EMT), cancer cell migration, and invasion. Targeting TGF-β signaling is a promising therapeutic approach, but nonspecific inhibition may result in adverse effects. In this study, we focus on the Smad2/3-Smad4 complex, a key component in TGF-β signaling transduction, as a potential target for cancer therapy. Through a phase-separated condensate-aided biomolecular interaction system, we identified verteporfin (VP) as a small-molecule inhibitor that specifically targets the Smad2/3-Smad4 interaction. VP effectively disrupted the interaction between Smad2/3 and Smad4 and thereby inhibited canonical TGF-β signaling, but not the interaction between Smad1 and Smad4 in bone morphogenetic protein (BMP) signaling. Furthermore, VP exhibited inhibitory effects on TGF-β-induced EMT and cell migration. Our findings indicate a novel approach to develop protein-protein interaction inhibitors of the canonical TGF-β signaling pathway for treatments of related diseases.
Collapse
Affiliation(s)
- Junxiu Nong
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shengqiang Shen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Fan Hong
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Fan Xiao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Lingtian Meng
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Pilong Li
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
- School of Basic Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
13
|
Liu Y, Dong L, Ma J, Chen L, Fang L, Wang Z. The prognostic genes model of breast cancer drug resistance based on single-cell sequencing analysis and transcriptome analysis. Clin Exp Med 2024; 24:113. [PMID: 38795164 PMCID: PMC11127859 DOI: 10.1007/s10238-024-01372-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/08/2024] [Indexed: 05/27/2024]
Abstract
Breast cancer (BC) represents a multifaceted malignancy, with escalating incidence and mortality rates annually. Chemotherapy stands as an indispensable approach for treating breast cancer, yet drug resistance poses a formidable challenge. Through transcriptome data analysis, we have identified two sets of genes exhibiting differential expression in this context. Furthermore, we have confirmed the overlap between these genes and those associated with exosomes, which were subsequently validated in cell lines. The investigation screened the identified genes to determine prognostic markers for BC and utilized them to formulate a prognostic model. The disparities in prognosis and immunity between the high- and low-risk groups were validated using the test dataset. We have discerned different BC subtypes based on the expression levels of prognostic genes in BC samples. Variations in prognosis, immunity, and drug sensitivity among distinct subtypes were examined. Leveraging data from single-cell sequencing and prognostic gene expression, the AUCell algorithm was employed to score individual cell clusters and analyze the pathways implicated in high-scoring groups. Prognostic genes (CCT4, CXCL13, MTDH, PSMD2, and RAB27A) were subsewoquently validated using RT-qPCR. Consequently, we have established a model for predicting prognosis in breast cancer that hinges on drug resistance and ERGs. Furthermore, we have evaluated the prognostic value of this model. The genes identified as prognostic markers can now serve as a reference for precise treatment of this condition.
Collapse
Affiliation(s)
- Yao Liu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Lun Dong
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jing Ma
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Linghui Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Liaoqiong Fang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
- National Engineering Research Center of Ultrasound Medicine, Chongqing, 401121, China.
| | - Zhibiao Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
- National Engineering Research Center of Ultrasound Medicine, Chongqing, 401121, China.
| |
Collapse
|
14
|
Khorami-Sarvestani S, Vanaki N, Shojaeian S, Zarnani K, Stensballe A, Jeddi-Tehrani M, Zarnani AH. Placenta: an old organ with new functions. Front Immunol 2024; 15:1385762. [PMID: 38707901 PMCID: PMC11066266 DOI: 10.3389/fimmu.2024.1385762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
The transition from oviparity to viviparity and the establishment of feto-maternal communications introduced the placenta as the major anatomical site to provide nutrients, gases, and hormones to the developing fetus. The placenta has endocrine functions, orchestrates maternal adaptations to pregnancy at different periods of pregnancy, and acts as a selective barrier to minimize exposure of developing fetus to xenobiotics, pathogens, and parasites. Despite the fact that this ancient organ is central for establishment of a normal pregnancy in eutherians, the placenta remains one of the least studied organs. The first step of pregnancy, embryo implantation, is finely regulated by the trophoectoderm, the precursor of all trophoblast cells. There is a bidirectional communication between placenta and endometrium leading to decidualization, a critical step for maintenance of pregnancy. There are three-direction interactions between the placenta, maternal immune cells, and the endometrium for adaptation of endometrial immune system to the allogeneic fetus. While 65% of all systemically expressed human proteins have been found in the placenta tissues, it expresses numerous placenta-specific proteins, whose expression are dramatically changed in gestational diseases and could serve as biomarkers for early detection of gestational diseases. Surprisingly, placentation and carcinogenesis exhibit numerous shared features in metabolism and cell behavior, proteins and molecular signatures, signaling pathways, and tissue microenvironment, which proposes the concept of "cancer as ectopic trophoblastic cells". By extensive researches in this novel field, a handful of cancer biomarkers has been discovered. This review paper, which has been inspired in part by our extensive experiences during the past couple of years, highlights new aspects of placental functions with emphasis on its immunomodulatory role in establishment of a successful pregnancy and on a potential link between placentation and carcinogenesis.
Collapse
Affiliation(s)
- Sara Khorami-Sarvestani
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Negar Vanaki
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sorour Shojaeian
- Department of Biochemistry, School of Medical Sciences, Alborz University of Medical Sciences, Karaj, Iran
| | - Kayhan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Han X, Song X, Xiao Z, Zhu G, Gao R, Ni B, Li J. Study on the mechanism of MDSC-platelets and their role in the breast cancer microenvironment. Front Cell Dev Biol 2024; 12:1310442. [PMID: 38404689 PMCID: PMC10884319 DOI: 10.3389/fcell.2024.1310442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are key immunosuppressive cells in the tumor microenvironment (TME) that play critical roles in promoting tumor growth and metastasis. Tumor-associated platelets (TAPs) help cancer cells evade the immune system and promote metastasis. In this paper, we describe the interaction between MDSCs and TAPs, including their generation, secretion, activation, and recruitment, as well as the effects of MDSCs and platelets on the generation and changes in the immune, metabolic, and angiogenic breast cancer (BC) microenvironments. In addition, we summarize preclinical and clinical studies, traditional Chinese medicine (TCM) therapeutic approaches, and new technologies related to targeting and preventing MDSCs from interacting with TAPs to modulate the BC TME, discuss the potential mechanisms, and provide perspectives for future development. The therapeutic strategies discussed in this review may have implications in promoting the normalization of the BC TME, reducing primary tumor growth and distant lung metastasis, and improving the efficiency of anti-tumor therapy, thereby improving the overall survival (OS) and progression-free survival (PFS) of patients. However, despite the significant advances in understanding these mechanisms and therapeutic strategies, the complexity and heterogeneity of MDSCs and side effects of antiplatelet agents remain challenging. This requires further investigation in future prospective cohort studies.
Collapse
Affiliation(s)
- Xinpu Han
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Hematology-Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotong Song
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhigang Xiao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guanghui Zhu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruike Gao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoyi Ni
- Department of Oncology, First Hospital of Heilongjiang University of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jie Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
16
|
Wang H, Wang X, Xu L. Transforming growth factor-induced gene TGFBI is correlated with the prognosis and immune infiltrations of breast cancer. World J Surg Oncol 2024; 22:22. [PMID: 38245723 PMCID: PMC10799375 DOI: 10.1186/s12957-024-03301-z] [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: 09/18/2023] [Accepted: 01/13/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Transforming growth factor β (TGFβ) is a critical regulator of lung metastasis of breast cancer and is correlated with the prognosis of breast cancer. However, not all TGFβ stimulated genes were functional and prognostic in breast cancer lung metastatic progress. In this study, we tried to determine the prognosis of TGFβ stimulated genes in breast cancer. METHODS TGFβ stimulated genes in MDA-MB-231 cells and lung metastasis-associated genes in LM2-4175 cells were identified through gene expression microarray. The prognosis of the induced gene (TGFBI) in breast cancer was determined through bioinformatics analysis and validated using tissue microarray. The immune infiltrations of breast cancer were determined through "ESTIMATE" and "TIMER". RESULTS TGFBI was up-regulated by TGFβ treatment and over-expressed in LM2-4175 cells. Through bioinformatics analysis, we found that higher expression of TGFBI was associated with shorted lung metastasis-free survival, relapse-free survival, disease-free survival, and overall survival of breast cancer. Moreover, the prognosis of TGFBI was validated in 139 Chinese breast cancer patients. Chinese breast cancer patients with higher TGFBI expression had lower overall survival. Correspondingly, breast cancer patients with higher TGFBI methylation had higher overall survival. TGFBI was correlated with the score of the TGFβ signaling pathway and multiple immune-related signaling pathways in breast cancer. The stromal score, immune score, and the infiltrations of immune cells were also correlated with TGFBI expression in breast cancer. CONCLUSIONS TGFβ-induced gene TGFBI was correlated with the prognosis and immune infiltrations of breast cancer.
Collapse
Affiliation(s)
- Haiwei Wang
- Medical Genetic Diagnosis and Therapy Center, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| | - Xinrui Wang
- Medical Genetic Diagnosis and Therapy Center, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Liangpu Xu
- Medical Genetic Diagnosis and Therapy Center, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
17
|
Yan S, Wang J, Chen H, Zhang D, Imam M. Divergent features of ERβ isoforms in triple negative breast cancer: progress and implications for further research. Front Cell Dev Biol 2023; 11:1240386. [PMID: 37936981 PMCID: PMC10626554 DOI: 10.3389/fcell.2023.1240386] [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: 06/14/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023] Open
Abstract
Estrogen receptor β (ERβ) was discovered more than 20 years ago. However, the extent and role of ERβ expression in breast cancer remain controversial, especially in the context of triple-negative breast cancer (TNBC). ERβ exists as multiple isoforms, and a series of studies has revealed an inconsistent role of ERβ isoforms in TNBC. Our recent results demonstrated contrasting functions of ERβ1 and ERβ2/β5 in TNBC. Additional research should be conducted to explore the functions of individual ERβ isoforms and develop targeted drugs according to the relevant mechanisms. Consequently, a systematic review of ERβ isoforms is necessary. In this review, we overview the structure of ERβ isoforms and detail what is known about the function of ERβ isoforms in normal mammary tissue and breast cancer. Moreover, this review highlights the divergent features of ERβ isoforms in TNBC. This review also provides insights into the implications of targeting ERβ isoforms for clinical treatment. In conclusion, this review provides a framework delineating the roles and mechanisms of different ERβ isoforms in TNBC and sheds light on future directions for basic and clinical research.
Collapse
Affiliation(s)
- Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | | | | | | | | |
Collapse
|
18
|
Chang CM, Chang CC, Lam HYP, Peng SY, Lai YH, Hsiang BD, Liao YY, Hsu HJ, Jiang SJ. Therapeutic Peptide RF16 Derived from CXCL8 Inhibits MDA-MB-231 Cell Invasion and Metastasis. Int J Mol Sci 2023; 24:14029. [PMID: 37762330 PMCID: PMC10531501 DOI: 10.3390/ijms241814029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Interleukin (IL)-8 plays a vital role in regulating inflammation and breast cancer formation by activating CXCR1/2. We previously designed an antagonist peptide, (RF16), to inhibits the activation of downstream signaling pathways by competing with IL-8 in binding to CXCR1/2, thereby inhibiting IL-8-induced chemoattractant monocyte binding. To evaluate the effect of the RF16 peptide on breast cancer progression, triple-negative MDA-MB-231 and ER-positive MCF-7 breast cancer cells were used to investigate whether RF16 can inhibit the IL-8-induced breast cancer metastasis. Using growth, proliferation, and invasiveness assays, the results revealed that RF16 reduced cell proliferation, migration, and invasiveness in MDA-MB-231 cells. The RF16 peptide also regulated the protein and mRNA expressions of epithelial-mesenchymal transition (EMT) markers in IL-8-stimulated MDA-MB-231 cells. It also inhibited downstream IL-8 signaling and the IL-8-induced inflammatory response via the mitogen-activated protein kinase (MAPK) and Phosphoinositide 3-kinase (PI3K) pathways. In the xenograft tumor mouse model, RF16 synergistically reinforces the antitumor efficacy of docetaxel by improving mouse survival and retarding tumor growth. Our results indicate that RF16 significantly inhibited IL-8-stimulated cell growth, migration, and invasion in MDA-MB-231 breast cancer cells by blocking the activation of p38 and AKT cascades. It indicated that the RF16 peptide may serve as a new supplementary drug for breast cancer.
Collapse
Affiliation(s)
- Chun-Ming Chang
- Department of General Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
| | - Chun-Chun Chang
- Department of Laboratory Medicine, Hualien Tzu Chi Hospital, Hualien 97004, Taiwan;
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Ho Yin Pekkle Lam
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (H.Y.P.L.); (S.-Y.P.); (Y.-H.L.)
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Shih-Yi Peng
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (H.Y.P.L.); (S.-Y.P.); (Y.-H.L.)
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Yi-Hsuan Lai
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (H.Y.P.L.); (S.-Y.P.); (Y.-H.L.)
| | - Bi-Da Hsiang
- Department of Molecular Biology and Human Genetics, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Yu-Yi Liao
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Hao-Jen Hsu
- Department of Biomedical Sciences and Engineering, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Shinn-Jong Jiang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (H.Y.P.L.); (S.-Y.P.); (Y.-H.L.)
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| |
Collapse
|
19
|
Dai Y, Zhang X, Ou Y, Zou L, Zhang D, Yang Q, Qin Y, Du X, Li W, Yuan Z, Xiao Z, Wen Q. Anoikis resistance--protagonists of breast cancer cells survive and metastasize after ECM detachment. Cell Commun Signal 2023; 21:190. [PMID: 37537585 PMCID: PMC10399053 DOI: 10.1186/s12964-023-01183-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/04/2023] [Indexed: 08/05/2023] Open
Abstract
Breast cancer exhibits the highest global incidence among all tumor types. Regardless of the type of breast cancer, metastasis is a crucial cause of poor prognosis. Anoikis, a form of apoptosis initiated by cell detachment from the native environment, is an outside-in process commencing with the disruption of cytosolic connectors such as integrin-ECM and cadherin-cell. This disruption subsequently leads to intracellular cytoskeletal and signaling pathway alterations, ultimately activating caspases and initiating programmed cell death. Development of an anoikis-resistant phenotype is a critical initial step in tumor metastasis. Breast cancer employs a series of stromal alterations to suppress anoikis in cancer cells. Comprehensive investigation of anoikis resistance mechanisms can inform strategies for preventing and regressing metastatic breast cancer. The present review first outlines the physiological mechanisms of anoikis, elucidating the alterations in signaling pathways, cytoskeleton, and protein targets that transpire from the outside in upon adhesion loss in normal breast cells. The specific anoikis resistance mechanisms induced by pathological changes in various spatial structures during breast cancer development are also discussed. Additionally, the genetic loci of targets altered in the development of anoikis resistance in breast cancer, are summarized. Finally, the micro-RNAs and targeted drugs reported in the literature concerning anoikis are compiled, with keratocin being the most functionally comprehensive. Video Abstract.
Collapse
Affiliation(s)
- Yalan Dai
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Oncology, Garze Tibetan Autonomous Prefecture People's Hospital, Kangding, China
| | - Xinyi Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Yingjun Ou
- Clinical Medicine School, Southwest Medicial Univercity, Luzhou, China
- Orthopaedics, Garze Tibetan Autonomous Prefecture People's Hospital, Kangding, China
| | - Linglin Zou
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Duoli Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qingfan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Qin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiuju Du
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Li
- Southwest Medical University, Luzhou, China
| | | | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
20
|
Mgrditchian T, Brown-Clay J, Hoffmann C, Müller T, Filali L, Ockfen E, Mao X, Moreau F, Casellas CP, Kaoma T, Mittelbronn M, Thomas C. Actin cytoskeleton depolymerization increases matrix metalloproteinase gene expression in breast cancer cells by promoting translocation of cysteine-rich protein 2 to the nucleus. Front Cell Dev Biol 2023; 11:1100938. [PMID: 37266453 PMCID: PMC10229898 DOI: 10.3389/fcell.2023.1100938] [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: 11/17/2022] [Accepted: 04/21/2023] [Indexed: 06/03/2023] Open
Abstract
The actin cytoskeleton plays a critical role in cancer cell invasion and metastasis; however, the coordination of its multiple functions remains unclear. Actin dynamics in the cytoplasm control the formation of invadopodia, which are membrane protrusions that facilitate cancer cell invasion by focusing the secretion of extracellular matrix-degrading enzymes, including matrix metalloproteinases (MMPs). In this study, we investigated the nuclear role of cysteine-rich protein 2 (CRP2), a two LIM domain-containing F-actin-binding protein that we previously identified as a cytoskeletal component of invadopodia, in breast cancer cells. We found that F-actin depolymerization stimulates the translocation of CRP2 into the nucleus, resulting in an increase in the transcript levels of pro-invasive and pro-metastatic genes, including several members of the MMP gene family. We demonstrate that in the nucleus, CRP2 interacts with the transcription factor serum response factor (SRF), which is crucial for the expression of MMP-9 and MMP-13. Our data suggest that CRP2 and SRF cooperate to modulate of MMP expression levels. Furthermore, Kaplan-Meier analysis revealed a significant association between high-level expression of SRF and shorter overall survival and distant metastasis-free survival in breast cancer patients with a high CRP2 expression profile. Our findings suggest a model in which CRP2 mediates the coordination of cytoplasmic and nuclear processes driven by actin dynamics, ultimately resulting in the induction of invasive and metastatic behavior in breast cancer cells.
Collapse
Affiliation(s)
- Takouhie Mgrditchian
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Joshua Brown-Clay
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Céline Hoffmann
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Tanja Müller
- Department of Cancer Research, Luxembourg Centre of Neuropathology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Liza Filali
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Elena Ockfen
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Xianqing Mao
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Flora Moreau
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Carla Pou Casellas
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Tony Kaoma
- Bioinformatics Platform, Luxembourg, Luxembourg
| | - Michel Mittelbronn
- Department of Cancer Research, Luxembourg Centre of Neuropathology, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-surAlzette, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-surAlzette, Luxembourg
- Department of Life Science and Medicine (DLSM), University of Luxembourg, Esch-surAlzette, Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
| | - Clément Thomas
- Department of Cancer Research, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
21
|
Malla R, Marni R, Chakraborty A. Exploring the role of CD151 in the tumor immune microenvironment: Therapeutic and clinical perspectives. Biochim Biophys Acta Rev Cancer 2023; 1878:188898. [PMID: 37094754 DOI: 10.1016/j.bbcan.2023.188898] [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: 02/11/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 04/26/2023]
Abstract
CD151 is a transmembrane protein implicated in tumor progression and has been shown to regulate various cellular and molecular mechanisms contributing to malignancy. More recently, the role of CD151 in the tumor immune microenvironment (TIME) has gained attention as a potential target for cancer therapy. This review aims to explore the role of CD151 in the TIME, focusing on the therapeutic and clinical perspectives. The role of CD151 in regulating the interactions between tumor cells and the immune system will be discussed, along with the current understanding of the molecular mechanisms underlying these interactions. The current state of the development of CD151-targeted therapies and the potential clinical applications of these therapies will also be reviewed. This review provides an overview of the current knowledge on the role of CD151 in the TIME and highlights the potential of CD151 as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Laboratory, Dept of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India.
| | - Rakshmita Marni
- Cancer Biology Laboratory, Dept of Biochemistry and Bioinformatics, GIS, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | | |
Collapse
|
22
|
Baldessari C, Pipitone S, Molinaro E, Cerma K, Fanelli M, Nasso C, Oltrecolli M, Pirola M, D’Agostino E, Pugliese G, Cerri S, Vitale MG, Madeo B, Dominici M, Sabbatini R. Bone Metastases and Health in Prostate Cancer: From Pathophysiology to Clinical Implications. Cancers (Basel) 2023; 15:1518. [PMID: 36900309 PMCID: PMC10000416 DOI: 10.3390/cancers15051518] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/15/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Clinically relevant bone metastases are a major cause of morbidity and mortality for prostate cancer patients. Distinct phenotypes are described: osteoblastic, the more common osteolytic and mixed. A molecular classification has been also proposed. Bone metastases start with the tropism of cancer cells to the bone through different multi-step tumor-host interactions, as described by the "metastatic cascade" model. Understanding these mechanisms, although far from being fully elucidated, could offer several potential targets for prevention and therapy. Moreover, the prognosis of patients is markedly influenced by skeletal-related events. They can be correlated not only with bone metastases, but also with "bad" bone health. There is a close correlation between osteoporosis-a skeletal disorder with decreased bone mass and qualitative alterations-and prostate cancer, in particular when treated with androgen deprivation therapy, a milestone in its treatment. Systemic treatments for prostate cancer, especially with the newest options, have improved the survival and quality of life of patients with respect to skeletal-related events; however, all patients should be evaluated for "bone health" and osteoporotic risk, both in the presence and in the absence of bone metastases. Treatment with bone-targeted therapies should be evaluated even in the absence of bone metastases, as described in special guidelines and according to a multidisciplinary evaluation.
Collapse
Affiliation(s)
- Cinzia Baldessari
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
| | - Stefania Pipitone
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
| | - Eleonora Molinaro
- Oncology, AUSL of Modena Area Sud, Sassuolo-Vignola-Pavullo, 41121 Modena, Italy
| | - Krisida Cerma
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology IOV—IRCCS, 35128 Padova, Italy
| | - Martina Fanelli
- Department of Oncology, Azienda Ospedaliero Universitaria S. M. della Misericordia, 33100 Udine, Italy
| | - Cecilia Nasso
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
- Medical Oncology, Ospedale Santa Corona, 17027 Pietra Ligure, Italy
| | - Marco Oltrecolli
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
| | - Marta Pirola
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
| | - Elisa D’Agostino
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
| | - Giuseppe Pugliese
- Department of Oncology and Hematology, Univerity of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Sara Cerri
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
| | - Maria Giuseppa Vitale
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
| | - Bruno Madeo
- Unit of Endocrinology, Department of Medical Specialities, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
| | - Massimo Dominici
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
| | - Roberto Sabbatini
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria of Modena, 41125 Modena, Italy
| |
Collapse
|
23
|
Organotropism of breast cancer metastasis: A comprehensive approach to the shared gene network. GENE REPORTS 2023. [DOI: 10.1016/j.genrep.2023.101749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
24
|
TOR1B: a predictor of bone metastasis in breast cancer patients. Sci Rep 2023; 13:1495. [PMID: 36707670 PMCID: PMC9883392 DOI: 10.1038/s41598-023-28140-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Recent therapeutic advances in breast cancer (BC) have improved survival outcomes; however, the prognosis for patients with bone metastasis (BM) remains poor. Hence, novel clinical biomarkers are needed to accurately predict BC BM as well as to promote personalized medicine. Here, we discovered a novel biomarker, TOR1B, for BM in BC patients via analysis of BC gene expression data and clinical information downloaded from open public databases. In cancer cells, we found high expression levels of TOR1B in the nucleus and endoplasmic reticulum. Regarding gene expression, the level of TOR1B was significantly upregulated in BC patients with BM (p < 0.05), and the result was externally validated. In addition, gene expression clearly demonstrated two distinct types of prognoses in ER- and PR-positive patients. In multivariate regression, the gene could be an independent predictor of BM in BC patients, i.e., a low expression level of TOR1B was associated with delayed metastasis to bone in BC patients (HR, 0.28; 95% CI 0.094-0.84). Conclusively, TOR1B might be a useful biomarker for predicting BM; specifically, patients with ER- and PR-positive subtypes would benefit from the clinical use of this promising prognostic biomarker.
Collapse
|
25
|
Tong X, Tang R, Xu J, Wang W, Zhao Y, Yu X, Shi S. Liquid-liquid phase separation in tumor biology. Signal Transduct Target Ther 2022; 7:221. [PMID: 35803926 PMCID: PMC9270353 DOI: 10.1038/s41392-022-01076-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 12/12/2022] Open
Abstract
Liquid-liquid phase separation (LLPS) is a novel principle for explaining the precise spatial and temporal regulation in living cells. LLPS compartmentalizes proteins and nucleic acids into micron-scale, liquid-like, membraneless bodies with specific functions, which were recently termed biomolecular condensates. Biomolecular condensates are executors underlying the intracellular spatiotemporal coordination of various biological activities, including chromatin organization, genomic stability, DNA damage response and repair, transcription, and signal transduction. Dysregulation of these cellular processes is a key event in the initiation and/or evolution of cancer, and emerging evidence has linked the formation and regulation of LLPS to malignant transformations in tumor biology. In this review, we comprehensively summarize the detailed mechanisms of biomolecular condensate formation and biophysical function and review the recent major advances toward elucidating the multiple mechanisms involved in cancer cell pathology driven by aberrant LLPS. In addition, we discuss the therapeutic perspectives of LLPS in cancer research and the most recently developed drug candidates targeting LLPS modulation that can be used to combat tumorigenesis.
Collapse
Affiliation(s)
- Xuhui Tong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rong Tang
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yingjun Zhao
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Si Shi
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
26
|
Nalla LV, Gondaliya P, Kalia K, Khairnar A. Targeting specificity protein 1 with miR-128-3p overcomes TGF-β1 mediated epithelial-mesenchymal transition in breast cancer: An in vitro study. Mol Biol Rep 2022; 49:6987-6996. [PMID: 35486287 DOI: 10.1007/s11033-022-07466-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/08/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Specificity protein 1 (SP1) was found to play a critical role in the regulation of TGF-β1 driven epithelial-mesenchymal transition (EMT). Recent clinical findings demonstrated a significant drop in the expression of miR-128-3p with the cancer progression in breast cancer patients. However, the impact of miR-128-3p on the SP1 expression in breast cancer remains unknown. Herein, we evaluated the role of miR-128-3p mimics in suppressing EMT of breast cancer cell lines by regulating the TGF-β1/SP1 axis. METHODS miR-128-3p interaction with SP1 was detected by in silico tools and dual-luciferase reporter assay. qPCR, western blot, and immunocytochemistry experiments were conducted for determining the expression levels of miR-128-3p and EMT markers with and without the treatment of miR-128-3p mimics. Further, to understand the effect of miR-128-3p mimics on cancer progression, experiments such as wound healing assay, transwell assay, adhesion assay, and cell cycle analysis were performed. RESULTS A significant inverse relation between SP1 and miR-128-3p levels was found in MCF-7 and MDA-MB-231 cell lines. miR-128-3p overexpression impeded the SP1 mediated EMT markers in TGF-β1 stimulated cells by inhibiting the SP1 nuclear function. Further, treatment with miR-128-3p mimics significantly reduced the migration, invasion and spreading capability of TGF-β1 stimulated cells. Flow cytometry results showed the impeding role of miR-128-3p on the cell cycle progression. CONCLUSIONS Upregulated miR-128-3p inhibited SP1, thereby limiting the TGF-β1 induced EMT in MCF-7 and MDA-MB-231 cell lines for the first time. This study may pave the path to explore novel miRNA therapeutics for eradicating advanced breast cancer cases.
Collapse
Affiliation(s)
- Lakshmi Vineela Nalla
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Palaj, Gandhinagar, Gujarat, 382355, India
| | - Piyush Gondaliya
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gandhinagar, Gujarat, 382355, India
| | - Kiran Kalia
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gandhinagar, Gujarat, 382355, India
| | - Amit Khairnar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
27
|
Plasma profile of immune determinants predicts pathological complete response in locally advanced breast cancer patients: a pilot study. Clin Breast Cancer 2022; 22:705-714. [DOI: 10.1016/j.clbc.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: 10/29/2021] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 11/24/2022]
|
28
|
Pruteanu LL, Braicu C, Módos D, Jurj MA, Raduly LZ, Zănoagă O, Magdo L, Cojocneanu R, Paşca S, Moldovan C, Moldovan AI, Ţigu AB, Gurzău E, Jäntschi L, Bender A, Berindan-Neagoe I. Targeting Cell Death Mechanism Specifically in Triple Negative Breast Cancer Cell Lines. Int J Mol Sci 2022; 23:ijms23094784. [PMID: 35563174 PMCID: PMC9099741 DOI: 10.3390/ijms23094784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022] Open
Abstract
Triple negative breast cancer (TNBC) is currently associated with a lack of treatment options. Arsenic derivatives have shown antitumoral activity both in vitro and in vivo; however, their mode of action is not completely understood. In this work we evaluate the response to arsenate of the double positive MCF-7 breast cancer cell line as well as of two different TNBC cell lines, Hs578T and MDA-MB-231. Multimodal experiments were conducted to this end, using functional assays and microarrays. Arsenate was found to induce cytoskeletal alteration, autophagy and apoptosis in TNBC cells, and moderate effects in MCF-7 cells. Gene expression analysis showed that the TNBC cell lines’ response to arsenate was more prominent in the G2M checkpoint, autophagy and apoptosis compared to the Human Mammary Epithelial Cells (HMEC) and MCF-7 cell lines. We confirmed the downregulation of anti-apoptotic genes (MCL1, BCL2, TGFβ1 and CCND1) by qRT-PCR, and on the protein level, for TGFβ2, by ELISA. Insight into the mode of action of arsenate in TNBC cell lines it is provided, and we concluded that TNBC and non-TNBC cell lines reacted differently to arsenate treatment in this particular experimental setup. We suggest the future research of arsenate as a treatment strategy against TNBC.
Collapse
Affiliation(s)
- Lavinia-Lorena Pruteanu
- Department of Chemistry, Centre for Molecular Science Informatics, University of Cambridge, Cambridge CB2 1EW, UK; (L.-L.P.); (D.M.); (A.B.)
- MedFuture Research Center for Advanced Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400377 Cluj-Napoca, Romania; (C.M.); (A.I.M.); (A.B.Ț.)
- Department of Chemistry and Biology, North University Center at Baia Mare, Technical University of Cluj-Napoca, 4800 Baia Mare, Romania
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (M.-A.J.); (L.-Z.R.); (O.Z.); (L.M.); (R.C.); (S.P.); (I.B.-N.)
- Correspondence:
| | - Dezső Módos
- Department of Chemistry, Centre for Molecular Science Informatics, University of Cambridge, Cambridge CB2 1EW, UK; (L.-L.P.); (D.M.); (A.B.)
| | - Maria-Ancuţa Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (M.-A.J.); (L.-Z.R.); (O.Z.); (L.M.); (R.C.); (S.P.); (I.B.-N.)
| | - Lajos-Zsolt Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (M.-A.J.); (L.-Z.R.); (O.Z.); (L.M.); (R.C.); (S.P.); (I.B.-N.)
| | - Oana Zănoagă
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (M.-A.J.); (L.-Z.R.); (O.Z.); (L.M.); (R.C.); (S.P.); (I.B.-N.)
| | - Lorand Magdo
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (M.-A.J.); (L.-Z.R.); (O.Z.); (L.M.); (R.C.); (S.P.); (I.B.-N.)
| | - Roxana Cojocneanu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (M.-A.J.); (L.-Z.R.); (O.Z.); (L.M.); (R.C.); (S.P.); (I.B.-N.)
| | - Sergiu Paşca
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (M.-A.J.); (L.-Z.R.); (O.Z.); (L.M.); (R.C.); (S.P.); (I.B.-N.)
| | - Cristian Moldovan
- MedFuture Research Center for Advanced Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400377 Cluj-Napoca, Romania; (C.M.); (A.I.M.); (A.B.Ț.)
- Department of Pharmaceutical Physics-Biophysics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alin Iulian Moldovan
- MedFuture Research Center for Advanced Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400377 Cluj-Napoca, Romania; (C.M.); (A.I.M.); (A.B.Ț.)
- Department of Pharmaceutical Physics-Biophysics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Adrian Bogdan Ţigu
- MedFuture Research Center for Advanced Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400377 Cluj-Napoca, Romania; (C.M.); (A.I.M.); (A.B.Ț.)
| | - Eugen Gurzău
- Environmental Health Center, 400240 Cluj-Napoca, Romania;
| | - Lorentz Jäntschi
- Institute for Doctoral Studies, Babeş-Bolyai University, 400084 Cluj-Napoca, Romania;
- Department of Physics and Chemistry, Technical University of Cluj-Napoca, 400641 Cluj-Napoca, Romania
| | - Andreas Bender
- Department of Chemistry, Centre for Molecular Science Informatics, University of Cambridge, Cambridge CB2 1EW, UK; (L.-L.P.); (D.M.); (A.B.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (M.-A.J.); (L.-Z.R.); (O.Z.); (L.M.); (R.C.); (S.P.); (I.B.-N.)
| |
Collapse
|
29
|
Abstract
OBJECTIVES Bone metastases are of high clinical relevance because they are a frequent complication of most types of common cancers, such as breast and prostate. The metastatic process is complex, requiring the completion of several different steps to allow successful dissemination and homing. In addition, preparation of the metastatic niche changes the constant cycle of bone matrix formation and degradation, leading to the clinical phenotypes of lytic and sclerotic lesions. We review our current knowledge on this topic and briefly explain the current treatment landscape of bone metastasis. DATA SOURCES These include PubMed, international guidelines, and clinician experience. CONCLUSION Bone metastases remain a clinical challenge that negatively impacts patients prognosis and quality of life. A comprehensive understanding of the complex molecular mechanisms that results in bone metastasis is the basis for successful treatment of affected patients. The disruption of bone matrix metabolism is already recognized as the prerequisite for metastasis formation, but many open questions remain that need to be addressed in future research to establish individually tailored treatment approaches. IMPLICATIONS FOR NURSING PRACTICE Patient-centered therapy of bone metastases requires suitable pharmacological options, and importantly a holistic approach in care delivery across the multidisciplinary team. Nurses provide the cornerstone of the multidisciplinary team and provide the closest and the most frequent contact to the patient and their families to provide timely intervention. Nurses require a basic understanding of the complex physiology of metastasis to inform practice.
Collapse
Affiliation(s)
- Romy M Riffel
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Andy Göbel
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Tilman D Rachner
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
30
|
Xiong Q, Zhang N, Zhang M, Wang M, Wang L, Fan Y, Lin CY. Engineer a pre-metastatic niched microenvironment to attract breast cancer cells by utilizing a 3D printed polycaprolactone/nano-hydroxyapatite osteogenic scaffold - An in vitro model system for proof of concept. J Biomed Mater Res B Appl Biomater 2022; 110:1604-1614. [PMID: 35112785 DOI: 10.1002/jbm.b.35021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 11/18/2021] [Accepted: 01/15/2022] [Indexed: 01/17/2023]
Abstract
Breast cancer bone metastasis is not a random process. It is affected by the local microenvironment which determines the propensity of cancer cells to invade and colonize into the secondary sites. This microenvironment is termed a pre-metastatic niche. With the flexibility to incorporate different biofactors, tissue-engineering scaffolds provide an advantageous environment to promote "designed" osteogenesis that may mimic the bony pre-metastatic niche. In the current study, designed polycaprolactone (PCL) scaffolds enriched with nano-hydroxyapatite (nHA) were fabricated through three-dimensional (3D) printing. Subsequently, human mesenchymal stem cells (hMSCs) were seeded onto PCL-nHA scaffolds for osteogenic differentiation to establish the pre-metastatic niched microenvironment. Furthermore, transwell migration assay was used to investigate recruitment of MDA-MB-231, MCF-7, and MDA-MB-453 breast cancer cells to the osseous PCL-nHA scaffolds. Our results showed that the mRNA levels of alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), and osteocalcin (OCN) of hMSCs on the PCL-nHA scaffolds were dramatically increased compared those with the PCL scaffolds (control) at day 7, 14, and 28. Meanwhile, the migration analysis showed that the higher maturation of osteogenesis and bone metabolism collectively contributed to the creation of a more favorable niched site for the cancerous invasion. Moreover, one of the hypothesized key mediators for the promoted migration, CXCL12, was confirmed using an assay of antagonist LIT-927. This early study demonstrated that a designed tissue engineering scaffold can be utilized to create a bone-mimicking environment that serves as a novel platform to recapitulate the pre-metastatic niche and help interrogate the scheme of bone metastasis by breast cancer.
Collapse
Affiliation(s)
- Qisheng Xiong
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Ningze Zhang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Miaomiao Zhang
- Beijing Institute of 3D Printing, Beijing City University, Beijing, China
| | - Meng Wang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Lizhen Wang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yubo Fan
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Chia-Ying Lin
- Department of Orthopaedic Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
31
|
Frankman ZD, Jiang L, Schroeder JA, Zohar Y. Application of Microfluidic Systems for Breast Cancer Research. MICROMACHINES 2022; 13:152. [PMID: 35208277 PMCID: PMC8877872 DOI: 10.3390/mi13020152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023]
Abstract
Cancer is a disease in which cells in the body grow out of control; breast cancer is the most common cancer in women in the United States. Due to early screening and advancements in therapeutic interventions, deaths from breast cancer have declined over time, although breast cancer remains the second leading cause of cancer death among women. Most deaths are due to metastasis, as cancer cells from the primary tumor in the breast form secondary tumors in remote sites in distant organs. Over many years, the basic biological mechanisms of breast cancer initiation and progression, as well as the subsequent metastatic cascade, have been studied using cell cultures and animal models. These models, although extremely useful for delineating cellular mechanisms, are poor predictors of physiological responses, primarily due to lack of proper microenvironments. In the last decade, microfluidics has emerged as a technology that could lead to a paradigm shift in breast cancer research. With the introduction of the organ-on-a-chip concept, microfluidic-based systems have been developed to reconstitute the dominant functions of several organs. These systems enable the construction of 3D cellular co-cultures mimicking in vivo tissue-level microenvironments, including that of breast cancer. Several reviews have been presented focusing on breast cancer formation, growth and metastasis, including invasion, intravasation, and extravasation. In this review, realizing that breast cancer can recur decades following post-treatment disease-free survival, we expand the discussion to account for microfluidic applications in the important areas of breast cancer detection, dormancy, and therapeutic development. It appears that, in the future, the role of microfluidics will only increase in the effort to eradicate breast cancer.
Collapse
Affiliation(s)
- Zachary D. Frankman
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85721, USA;
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA;
| | - Joyce A. Schroeder
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA;
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA;
| |
Collapse
|
32
|
Li BB, Scott EY, Olafsen NE, Matthews J, Wheeler AR. Analysis of the effects of aryl hydrocarbon receptor expression on cancer cell invasion via three-dimensional microfluidic invasion assays. LAB ON A CHIP 2022; 22:313-325. [PMID: 34904612 DOI: 10.1039/d1lc00854d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that binds to xenobiotics and activates expression of response elements to metabolize these compounds. The AHR pathway has been associated with a long list of diseases including cancer; however, it is debated whether AHR is tumorigenic or tumour-inhibiting. In particular, there are contradictory reports in the literature regarding the effects of AHR expression level on metastatic breast cancer. Here we used a 3D invasion assay called cell invasion in digital microfluidic microgel systems (CIMMS) to study the effect of AHR expression on invasion. In this study, MDA-MB-231 cells with stable knockout of AHR (AHRko) showed enhanced invasive characteristics and reduced proliferation, and cells with transient overexpression of AHR showed reduced invasiveness. Overexpression of AHR with a mutation in the DNA binding domain showed no difference in invasiveness compared to control, which suggests that the changes in invasiveness are related to the expression of AHR. CIMMS also allowed for extraction of sub-populations of invaded cells for RNA sequencing experiments. A comparison of the transcriptomes of invaded subpopulations of wild-type and AHRko cells identified 1809 genes that were differentially expressed, with enriched pathways including cell cycle, proliferation, survival, immunoproteasome activation, and activation of matrix metalloproteases. In sum, the data reported here for MDA-MB-231 cells suggests some new interpretations of the discrepancy in the literature on the role of AHR in breast cancer. We propose that the unique combination of functional discrimination with transcriptome profiling provided by CIMMS will be valuable for a wide range of mechanistic invasion-biology studies in the future.
Collapse
Affiliation(s)
- Bingyu B Li
- Institute of Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON, M5S 3G9, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
| | - Erica Y Scott
- Institute of Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON, M5S 3G9, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
- Department of Chemistry, University of Toronto, 80 St George St., Toronto, ON, M5S 3H6, Canada
| | - Ninni E Olafsen
- Institute of Basic Medical Sciences, University of Oslo, Oslo, 0317, Norway
| | - Jason Matthews
- Institute of Basic Medical Sciences, University of Oslo, Oslo, 0317, Norway
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Aaron R Wheeler
- Institute of Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON, M5S 3G9, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
- Department of Chemistry, University of Toronto, 80 St George St., Toronto, ON, M5S 3H6, Canada
| |
Collapse
|
33
|
Mondal M, Conole D, Nautiyal J, Tate EW. UCHL1 as a novel target in breast cancer: emerging insights from cell and chemical biology. Br J Cancer 2022; 126:24-33. [PMID: 34497382 PMCID: PMC8727673 DOI: 10.1038/s41416-021-01516-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 06/25/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer has the highest incidence and death rate among cancers in women worldwide. In particular, metastatic estrogen receptor negative (ER-) breast cancer and triple-negative breast cancer (TNBC) subtypes have very limited treatment options, with low survival rates. Ubiquitin carboxyl terminal hydrolase L1 (UCHL1), a ubiquitin C-terminal hydrolase belonging to the deubiquitinase (DUB) family of enzymes, is highly expressed in these cancer types, and several key reports have revealed emerging and important roles for UCHL1 in breast cancer. However, selective and potent small-molecule UCHL1 inhibitors have been disclosed only very recently, alongside chemical biology approaches to detect regulated UHCL1 activity in cancer cells. These tools will enable novel insights into oncogenic mechanisms driven by UCHL1, and identification of substrate proteins deubiquitinated by UCHL1, with the ultimate goal of realising the potential of UCHL1 as a drug target in breast cancer.
Collapse
Affiliation(s)
- Milon Mondal
- Department of Chemistry, Imperial College London, London, UK
| | - Daniel Conole
- Department of Chemistry, Imperial College London, London, UK
| | - Jaya Nautiyal
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, London, UK.
| |
Collapse
|
34
|
Xiong Q, Wang M, Liu J, Lin CY. Breast Cancer Cells Metastasize to the Tissue-Engineered Premetastatic Niche by Using an Osteoid-Formed Polycaprolactone/Nanohydroxyapatite Scaffold. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:9354202. [PMID: 34938359 PMCID: PMC8687766 DOI: 10.1155/2021/9354202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/23/2021] [Indexed: 11/21/2022]
Abstract
It has been deemed that the premetastatic niche (PMN) plays a critical role in facilitating bone metastasis of breast cancer cells. Tissue engineering scaffolds provide an advantageous environment to promote osteogenesis that may mimic the bony premetastatic niches (BPMNs). In this study, human mesenchymal stem cells (hMSCs) were seeded onto designed polycaprolactone/nanohydroxyapatite (PCL-nHA) scaffolds for osteogenic differentiation. Subsequently, a coculture system was used to establish the tissue-engineered BPMNs by culturing breast cancer cells, hMSCs, and osteoid-formed PCL-nHA scaffolds. Afterwards, a migration assay was used to investigate the recruitment of MDA-MB-231, MCF-7, and MDA-MB-453 cells to the BPMNs' supernatants. The cancer stem cell (CSC) properties of these migrated cells were investigated by flow cytometry. Our results showed that the mRNA expression levels of alkaline phosphatase (ALP), Osterix, runt-related transcription factor 2 (Runx2), and collagen type I alpha 1 (COL1A1) on the PCL-nHA scaffolds were dramatically increased compared to the PCL scaffolds on days 11, 18, and 32. The expression of CXCL12 in these BPMNs was increased gradually over coculturing time, and it may be a feasible marker for BPMNs. Furthermore, migration analysis results showed that the higher maturation of BPMNs collectively contributed to the creation of a more favorable niched site for the cancerous invasion. The subpopulation of breast cancer stem cells (BCSCs) was more likely to migrate to fertile BPMNs. The proportion of BCSCs in metastatic MDA-MB-231, MCF-7, and MDA-MB-453 cells were increased by approximately 63.47%, 149.48%, and 127.60%. The current study demonstrated that a designed tissue engineering scaffold can provide a novel method to create a bone-mimicking environment that serves as a useable platform to recapitulate the BPMNs and help interrogate the scheme of bone metastasis by breast cancer.
Collapse
Affiliation(s)
- Qisheng Xiong
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Meng Wang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Jinglong Liu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Chia-Ying Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
- Department of Orthopaedic Surgery, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
35
|
Rodenburg WS, van Buul JD. Rho GTPase signalling networks in cancer cell transendothelial migration. VASCULAR BIOLOGY 2021; 3:R77-R95. [PMID: 34738075 PMCID: PMC8558887 DOI: 10.1530/vb-21-0008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/21/2023]
Abstract
Rho GTPases are small signalling G-proteins that are central regulators of cytoskeleton dynamics, and thereby regulate many cellular processes, including the shape, adhesion and migration of cells. As such, Rho GTPases are also essential for the invasive behaviour of cancer cells, and thus involved in several steps of the metastatic cascade, including the extravasation of cancer cells. Extravasation, the process by which cancer cells leave the circulation by transmigrating through the endothelium that lines capillary walls, is an essential step for metastasis towards distant organs. During extravasation, Rho GTPase signalling networks not only regulate the transmigration of cancer cells but also regulate the interactions between cancer and endothelial cells and are involved in the disruption of the endothelial barrier function, ultimately allowing cancer cells to extravasate into the underlying tissue and potentially form metastases. Thus, targeting Rho GTPase signalling networks in cancer may be an effective approach to inhibit extravasation and metastasis. In this review, the complex process of cancer cell extravasation will be discussed in detail. Additionally, the roles and regulation of Rho GTPase signalling networks during cancer cell extravasation will be discussed, both from a cancer cell and endothelial cell point of view.
Collapse
Affiliation(s)
- Wessel S Rodenburg
- Molecular Cell Biology Lab at Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab at Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands.,Leeuwenhoek Centre for Advanced Microscopy, Section Molecular Cytology at Swammerdam Institute for Life Sciences at University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
36
|
Use of relevancy and complementary information for discriminatory gene selection from high-dimensional gene expression data. PLoS One 2021; 16:e0230164. [PMID: 34613963 PMCID: PMC8494339 DOI: 10.1371/journal.pone.0230164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 09/21/2021] [Indexed: 12/22/2022] Open
Abstract
With the advent of high-throughput technologies, life sciences are generating a huge amount of varied biomolecular data. Global gene expression profiles provide a snapshot of all the genes that are transcribed in a cell or in a tissue under a particular condition. The high-dimensionality of such gene expression data (i.e., very large number of features/genes analyzed with relatively much less number of samples) makes it difficult to identify the key genes (biomarkers) that are truly attributing to a particular phenotype or condition, (such as cancer), de novo. For identifying the key genes from gene expression data, among the existing literature, mutual information (MI) is one of the most successful criteria. However, the correction of MI for finite sample is not taken into account in this regard. It is also important to incorporate dynamic discretization of genes for more relevant gene selection, although this is not considered in the available methods. Besides, it is usually suggested in current studies to remove redundant genes which is particularly inappropriate for biological data, as a group of genes may connect to each other for downstreaming proteins. Thus, despite being redundant, it is needed to add the genes which provide additional useful information for the disease. Addressing these issues, we proposed Mutual information based Gene Selection method (MGS) for selecting informative genes. Moreover, to rank these selected genes, we extended MGS and propose two ranking methods on the selected genes, such as MGSf—based on frequency and MGSrf—based on Random Forest. The proposed method not only obtained better classification rates on gene expression datasets derived from different gene expression studies compared to recently reported methods but also detected the key genes relevant to pathways with a causal relationship to the disease, which indicate that it will also able to find the responsible genes for an unknown disease data.
Collapse
|
37
|
A computational approach on studying the regulation of TGF-β1-stimulated Runx2 expression by MicroRNAs in human breast cancer cells. Comput Biol Med 2021; 137:104823. [PMID: 34492519 DOI: 10.1016/j.compbiomed.2021.104823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/15/2021] [Accepted: 08/29/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND Transforming growth factor-beta1 (TGF-β1) acts as a most effective growth inhibitor for normal epithelial cells. Loss of this anti-proliferative factor in breast tissues favors invasion and development of osteolytic metastases, aided by a master transcription factor, runt-related transcription factor 2 (Runx2). Several reports identified Runx2 regulation with the help of non-coding RNAs such as microRNAs (miRNAs) under physiological and pathological conditions. METHODS Using bioinformatics tools such as miRDB, STarMir, Venny, TarBase, a unique list of miRNAs that putatively target the 3' UTR Runx2 was identified. Further, the expression patterns of those miRNAs at the precursor and mature levels were studied by RT-qPCR analyses. Following this, computational analyses using software like TransmiR and bc-GenExMiner v4.6 were done to speculate the miRNA's other target genes that indirectly regulate Runx2 activity in breast cancer. RESULTS There were 13 miRNAs that putatively target Runx2 identified using bioinformatics tools. Among these miRNAs, miR-5703 expression was significantly downregulated at both precursor and mature levels upon TGF-β1-treatment in human breast cancer cells. Computational analyses speculated an indirect targeting of Runx2 by miR-5703 by influencing multiple Runx2 regulatory signaling pathways including Jak/Stat, MAPK, Wnt/β-Catenin, Notch, BMP, and PKA pathways. Furthermore, a correlation of the expression profiles of the speculated genes and Runx2 with miR-5703 was depicted in triple-negative breast cancer patients. CONCLUSION Identification of miR-5703 and its network for Runx2 regulation directly or indirectly in breast cancer cells could significantly advance our understanding of breast cancer-mediated bone metastasis. In addition, it would potentially pave the way for miRNAs to be used as biomarkers and therapeutic agents in cancer research.
Collapse
|
38
|
Argote Camacho AX, González Ramírez AR, Pérez Alonso AJ, Rejón García JD, Olivares Urbano MA, Torné Poyatos P, Ríos Arrabal S, Núñez MI. Metalloproteinases 1 and 3 as Potential Biomarkers in Breast Cancer Development. Int J Mol Sci 2021; 22:ijms22169012. [PMID: 34445715 PMCID: PMC8396449 DOI: 10.3390/ijms22169012] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer continues to be one of the main causes of morbidity and mortality globally and was the leading cause of cancer death in women in Spain in 2020. Early diagnosis is one of the most effective methods to lower the incidence and mortality rates of breast cancer. The human metalloproteinases (MMP) mainly function as proteolytic enzymes degrading the extracellular matrix and plays important roles in most steps of breast tumorigenesis. This retrospective cohort study shows the immunohistochemical expression levels of MMP-1, MMP-2, MMP-3, and MMP-9 in 154 women with breast cancer and 42 women without tumor disease. The samples of breast tissue are assessed using several tissue matrices (TMA). The percentages of staining (≤50%–>50%) and intensity levels of staining (weak, moderate, or intense) are considered. The immunohistochemical expression of the MMP-1-intensity (p = 0.043) and MMP-3 percentage (p = 0.018) and intensity, (p = 0.025) present statistically significant associations with the variable group (control–case); therefore, expression in the tumor tissue samples of these MMPs may be related to the development of breast cancer. The relationships between these MMPs and some clinicopathological factors in breast cancer are also evaluated but no correlation is found. These results suggest the use of MMP-1 and MMP-3 as potential biomarkers of breast cancer diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Pablo Torné Poyatos
- Department of Surgery and Its Specialties, University of Granada, 18012 Granada, Spain;
| | - Sandra Ríos Arrabal
- Department of Radiology and Physical Medicine, University of Granada, 18012 Granada, Spain;
- Correspondence: (S.R.A.); (M.I.N.); Tel.: +34-958-242077 (S.R.A.); +34-958-242077 (M.I.N.)
| | - María Isabel Núñez
- Department of Radiology and Physical Medicine, University of Granada, 18012 Granada, Spain;
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18016 Granada, Spain
- Biosanitary Research Institute, ibs.Granada, 18012 Granada, Spain
- Correspondence: (S.R.A.); (M.I.N.); Tel.: +34-958-242077 (S.R.A.); +34-958-242077 (M.I.N.)
| |
Collapse
|
39
|
Qiao X, Zhang Y, Sun L, Ma Q, Yang J, Ai L, Xue J, Chen G, Zhang H, Ji C, Gu X, Lei H, Yang Y, Liu C. Association of human breast cancer CD44 -/CD24 - cells with delayed distant metastasis. eLife 2021; 10:65418. [PMID: 34318746 PMCID: PMC8346282 DOI: 10.7554/elife.65418] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/25/2021] [Indexed: 12/09/2022] Open
Abstract
Tumor metastasis remains the main cause of breast cancer-related deaths, especially delayed breast cancer distant metastasis. The current study assessed the frequency of CD44-/CD24- breast cancer cells in 576 tissue specimens for associations with clinicopathological features and metastasis and investigated the underlying molecular mechanisms. The results indicated that higher frequency (≥19.5%) of CD44-/CD24- cells was associated with delayed postoperative breast cancer metastasis. Furthermore, CD44-/CD24-triple negative breast cancer (TNBC) cells spontaneously converted into CD44+/CD24-cancer stem cells (CSCs) with properties similar to CD44+/CD24-CSCs from primary human breast cancer cells and parental TNBC cells in terms of stemness marker expression, self-renewal, differentiation, tumorigenicity, and lung metastasis in vitro and in NOD/SCID mice. RNA sequencing identified several differentially expressed genes (DEGs) in newly converted CSCs and RHBDL2, one of the DEGs, expression was upregulated. More importantly, RHBDL2 silencing inhibited the YAP1/USP31/NF-κB signaling and attenuated spontaneous CD44-/CD24- cell conversion into CSCs and their mammosphere formation. These findings suggest that the frequency of CD44-/CD24- tumor cells and RHBDL2 may be valuable for prognosis of delayed breast cancer metastasis, particularly for TNBC.
Collapse
Affiliation(s)
- Xinbo Qiao
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yixiao Zhang
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China.,Dapartment of Urology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Lisha Sun
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Qingtian Ma
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Jie Yang
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Liping Ai
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Guanglei Chen
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Hao Zhang
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China.,Department of Breast Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Ce Ji
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China.,Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xi Gu
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Haixin Lei
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Yongliang Yang
- Center for Molecular Medicine, School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Caigang Liu
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
40
|
Göbel A, Dell’Endice S, Jaschke N, Pählig S, Shahid A, Hofbauer LC, Rachner TD. The Role of Inflammation in Breast and Prostate Cancer Metastasis to Bone. Int J Mol Sci 2021; 22:5078. [PMID: 34064859 PMCID: PMC8151893 DOI: 10.3390/ijms22105078] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor metastasis to bone is a common event in multiple forms of malignancy. Inflammation holds essential functions in homeostasis as a defense mechanism against infections and is a strategy to repair injured tissue and to adapt to stress conditions. However, exaggerated and/or persistent (chronic) inflammation may eventually become maladaptive and evoke diseases such as autoimmunity, diabetes, inflammatory tissue damage, fibrosis, and cancer. In fact, inflammation is now considered a hallmark of malignancy with prognostic relevance. Emerging studies have revealed a central involvement of inflammation in several steps of the metastatic cascade of bone-homing tumor cells through supporting their survival, migration, invasion, and growth. The mechanisms by which inflammation favors these steps involve activation of epithelial-to-mesenchymal transition (EMT), chemokine-mediated homing of tumor cells, local activation of osteoclastogenesis, and a positive feedback amplification of the protumorigenic inflammation loop between tumor and resident cells. In this review, we summarize established and evolving concepts of inflammation-driven tumorigenesis, with a special focus on bone metastasis.
Collapse
Affiliation(s)
- Andy Göbel
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Stefania Dell’Endice
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Nikolai Jaschke
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
- Center for Healthy Aging, Technische Universität Dresden, 01159 Dresden, Germany
| | - Sophie Pählig
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
| | - Amna Shahid
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
| | - Lorenz C. Hofbauer
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Healthy Aging, Technische Universität Dresden, 01159 Dresden, Germany
| | - Tilman D. Rachner
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Healthy Aging, Technische Universität Dresden, 01159 Dresden, Germany
| |
Collapse
|
41
|
Zhang X, Li X, Sun S, Wang P, Ma X, Hou R, Liang X. Anti-Tumor Metastasis via Platelet Inhibitor Combined with Photothermal Therapy under Activatable Fluorescence/Magnetic Resonance Bimodal Imaging Guidance. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19679-19694. [PMID: 33876926 DOI: 10.1021/acsami.1c02302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Photothermal therapy (PTT) is a promising tumor therapy strategy; however, heterogeneous heat distribution over the tumor often exists, resulting in insufficient photothermal ablation and potential risk of cancer metastasis, which has been demonstrated to be associate with platelets. Herein, a near-infrared (NIR) photothermal agent of IR780 was conjugated with MRI agent of Gd-DOTA via a disulfide linkage (ICD-Gd), which was coassembly with lipid connecting tumor-homing pentapeptide CREKA (Cys-Arg-Glu-Lys-Ala) (DSPE-PEG-CREKA) to encapsulate a platelet inhibitor of ticagrelor (Tic), affording a multistimuli-responsive nanosystem (DPC@ICD-Gd-Tic). The nanosystem with completely quenching fluorescence could specifically target the tumor-associated platelets and showed pH/reduction/NIR light-responsive drug release, which simultaneously resulting in dis-assembly of nanoparticle and fluorescence recovery, enabling the drug delivery visualization in tumor in situ via activatable NIR fluorescence/MR bimodal imaging. Finally, DPC@ICD-Gd-Tic further integrated the photoinduced hyperthermia and platelet function inhibitor to achieve synergistic anticancer therapy, leading to ablation of primary tumor cells and effectively suppressed their distant metastasis. The number of lung metastases in 4T1 tumor bearing mice was reduced by about 90%, and the size of tumor was reduced by about 70%, while half of the mouse was completely cured by this smart nanosystem.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, P. R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Xiaoda Li
- School of Basic Medical Sciences, Peking University, Beijing 100190, P. R. China
| | - Suhui Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, P. R. China
| | - Ping Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, P. R. China
| | - Xiaotu Ma
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, P. R. China
| | - Rui Hou
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, P. R. China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, P. R. China
| |
Collapse
|
42
|
Zhang MK, Shang QJ, Li SY, Wang B, Liu G, Wang ZL. TGF-β1: is it related to the stiffness of breast lesions and can it predict axillary lymph node metastasis? ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:870. [PMID: 34164504 PMCID: PMC8184473 DOI: 10.21037/atm-21-1705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background This study aimed to explore whether transforming growth factor β1 (TGF-β1) is correlated with the stiffness of breast lesions and if it can predict axillary lymph node (ALN) metastasis. Methods A retrospective analysis was performed in our hospital. A total of 135 breast lesions in 130 patients who were to undergo vacuum-assisted excisional biopsy (VAEB) or surgery were enrolled between April 2018 and October 2018. Ultrasound (US) and shear wave elastography (SWE) examinations were performed for every lesion before VAEB or surgery. Pathology results obtained by VAEB or surgery were regarded as gold criteria. The elastic parameters and TGF-β1 expression level of malignant breast lesions were compared with those of benign lesions; the relationship between TGF-β1 expression level in breast lesions and the elastic parameters was analyzed; the TGF-β1 expression level in breast lesions with or without ALN metastasis were compared; and the efficacy of TGF-β1 expression level in predicting ALN metastasis was analyzed. Results The malignant breast lesions were different from benign lesions in the maximum and mean elasticity (Emax, Emean), standard deviation of elasticity (ESD), elastic ratio of the lesions to the peripheral tissue (Eratio), and the occurrence rate of "stiff rim sign" (P<0.001). The expression level of TGF-β1 in benign breast lesions was significantly lower than that in malignant lesions (P<0.001), and the TGF-β1 expression level was positively correlated with Emax, Emean, ESD, and Eratio (r=0.869, 0.840, 0.834, and 0.734, respectively). The expression level of TGF-β1 in breast lesions with or without "stiff rim sign" was significantly different (P<0.001), and the TGF-β1 expression level in malignant breast lesions with ALN metastasis was significantly higher than that in malignant lesions without ALN metastasis (P=0.0009). When TGF-β1 expression level >0.3138 was taken as the cut-off value, its efficacy in predicting ALN metastasis was 0.853, with a sensitivity of 86.67%, and a specificity 83.33%. Conclusions The expression level of TGF-β1 was positively correlated with the elastic parameters of breast lesions, and it could be useful for predicting ALN metastasis, especially for negative ALN diagnosis clinically.
Collapse
Affiliation(s)
- Meng Ke Zhang
- Department of Ultrasound, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Qiu Jing Shang
- Department of Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Shi Yu Li
- Department of Ultrasound, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Bo Wang
- Department of Ultrasound, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Gang Liu
- Department of Radiology, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhi Li Wang
- Department of Ultrasound, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
43
|
Targeting Pin1 for Modulation of Cell Motility and Cancer Therapy. Biomedicines 2021; 9:biomedicines9040359. [PMID: 33807199 PMCID: PMC8065645 DOI: 10.3390/biomedicines9040359] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 01/09/2023] Open
Abstract
Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) specifically binds and isomerizes the phosphorylated serine/threonine-proline (pSer/Thr-Pro) motif, which leads to changes in protein conformation and function. Pin1 is widely overexpressed in cancers and plays an important role in tumorigenesis. Mounting evidence has revealed that targeting Pin1 is a potential therapeutic approach for various cancers by inhibiting cell proliferation, reducing metastasis, and maintaining genome stability. In this review, we summarize the underlying mechanisms of Pin1-mediated upregulation of oncogenes and downregulation of tumor suppressors in cancer development. Furthermore, we also discuss the multiple roles of Pin1 in cancer hallmarks and examine Pin1 as a desirable pharmaceutical target for cancer therapy. We also summarize the recent progress of Pin1-targeted small-molecule compounds for anticancer activity.
Collapse
|
44
|
Javed Z, Khan K, Rasheed A, Sadia H, Raza S, Salehi B, Cho WC, Sharifi-Rad J, Koch W, Kukula-Koch W, Głowniak-Lipa A, Helon P. MicroRNAs and Natural Compounds Mediated Regulation of TGF Signaling in Prostate Cancer. Front Pharmacol 2021; 11:613464. [PMID: 33584291 PMCID: PMC7873640 DOI: 10.3389/fphar.2020.613464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/24/2020] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer (PCa) is with rising incidence in male population globally. It is a complex anomaly orchestrated by a plethora of cellular processes. Transforming growth factor-beta (TGF-β) signaling is one of the key signaling pathways involved in the tumorigenesis of PCa. TGF-β signaling has a dual role in the PCa, making it difficult to find a suitable therapeutic option. MicroRNAs (miRNAs) mediated regulation of TGF-β signaling is responsible for the TGF-ß paradox. These are small molecules that modulate the expression of target genes and regulate cancer progression. Thus, miRNAs interaction with different signaling cascades is of great attention for devising new diagnostic and therapeutic options for PCa. Natural compounds have been extensively studied due to their high efficacy and low cytotoxicity. Here, we discuss the involvement of TGF-ß signaling in PCa with the interplay between miRNAs and TGF-β signaling and also review the role of natural compounds for the development of new therapeutics for PCa.
Collapse
Affiliation(s)
- Zeeshan Javed
- Office for Research Innovation and Commercialization, Lahore Garrison University, Lahore, Pakistan
| | - Khushbukhat Khan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Amna Rasheed
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Haleema Sadia
- Department of Biotechnology, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta, Pakistan
| | - Shahid Raza
- Office for Research Innovation and Commercialization, Lahore Garrison University, Lahore, Pakistan
| | - Bahare Salehi
- Medical Ethics and Law Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | - Wojciech Koch
- Chair and Department of Food and Nutrition, Medical University of Lublin, Lublin, Poland
| | | | - Anna Głowniak-Lipa
- Department of Cosmetology, University of Information Technology and Management in Rzeszów, Rzeszów, Poland
| | - Paweł Helon
- Branch in Sandomierz, Jan Kochanowski University in Kielce, Sandomierz, Poland
| |
Collapse
|
45
|
Micalizzi DS, Ebright RY, Haber DA, Maheswaran S. Translational Regulation of Cancer Metastasis. Cancer Res 2021; 81:517-524. [PMID: 33479028 DOI: 10.1158/0008-5472.can-20-2720] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/25/2020] [Accepted: 10/23/2020] [Indexed: 11/16/2022]
Abstract
Deregulation of the mRNA translational process has been observed during tumorigenesis. However, recent findings have shown that deregulation of translation also contributes specifically to cancer cell spread. During metastasis, cancer cells undergo changes in cellular state, permitting the acquisition of features necessary for cell survival, dissemination, and outgrowth. In addition, metastatic cells respond to external cues, allowing for their persistence under significant cellular and microenvironmental stresses. Recent work has revealed the importance of mRNA translation to these dynamic changes, including regulation of cell states through epithelial-to-mesenchymal transition and tumor dormancy and as a response to external stresses such as hypoxia and immune surveillance. In this review, we focus on examples of altered translation underlying these phenotypic changes and responses to external cues and explore how they contribute to metastatic progression. We also highlight the therapeutic opportunities presented by aberrant mRNA translation, suggesting novel ways to target metastatic tumor cells.
Collapse
Affiliation(s)
- Douglas S Micalizzi
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts.,Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Richard Y Ebright
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts. .,Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts. .,Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| |
Collapse
|
46
|
Maiti A, Hait NC. Autophagy-mediated tumor cell survival and progression of breast cancer metastasis to the brain. J Cancer 2021; 12:954-964. [PMID: 33442395 PMCID: PMC7797661 DOI: 10.7150/jca.50137] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/07/2020] [Indexed: 02/07/2023] Open
Abstract
Brain metastases represent a substantial amount of morbidity and mortality in breast cancer (BC). Metastatic breast tumor cells committed to brain metastases are unique because they escape immune surveillance, can penetrate the blood-brain barrier, and also adapt to the brain tissue microenvironment (TME) for colonization and outgrowth. In addition, dynamic intracellular interactions between metastatic cancer cells and neighboring astrocytes in the brain are thought to play essential roles in brain tumor progression. A better understanding of the above mechanisms will lead to developing more effective therapies for brain metastases. Growing literature suggests autophagy, a conserved lysosomal degradation pathway involved in cellular homeostasis under stressful conditions, plays essential roles in breast tumor metastatic transformation and brain metastases. Cancer cells must adapt under various microenvironmental stresses, such as hypoxia, and nutrient (glucose) deprivation, in order to survive and progress. Clinical studies reveal that tumoral expression of autophagy-related proteins is higher in brain metastasis compared to primary breast tumors. In this review, we outline the molecular mechanisms underlying autophagy-mediated BC cell survival and metastasis to the brain.
Collapse
Affiliation(s)
- Aparna Maiti
- Division of Breast Surgery and Department of Surgical Oncology, Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, 14263, USA
| | - Nitai C. Hait
- Division of Breast Surgery and Department of Surgical Oncology, Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, 14263, USA
| |
Collapse
|
47
|
Plavc G, Jesenko T, Oražem M, Strojan P. Challenges in Combining Immunotherapy with Radiotherapy in Recurrent/Metastatic Head and Neck Cancer. Cancers (Basel) 2020; 12:E3197. [PMID: 33143094 PMCID: PMC7692120 DOI: 10.3390/cancers12113197] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy with immune checkpoint inhibitors (ICI) has recently become a standard part of the treatment of recurrent or metastatic head and neck squamous cell carcinoma (R/M HNSCC), although the response rates are low. Numerous preclinical and clinical studies have now illuminated several mechanisms by which radiotherapy (RT) enhances the effect of ICI. From RT-induced immunogenic cancer cell death to its effect on the tumor microenvironment and vasculature, the involved mechanisms are diverse and intertwined. Moreover, the research of these interactions is challenging because of the thin line between immunostimulatory and the immunosuppressive effect of RT. In the era of active research of immunoradiotherapy combinations, the significance of treatment and host-related factors that were previously seen as being less important is being revealed. The impact of dose and fractionation of RT is now well established, whereas selection of the number and location of the lesions to be irradiated in a multi-metastatic setting is something that is only now beginning to be understood. In addition to spatial factors, the timing of irradiation is as equally important and is heavily dependent on the type of ICI used. Interestingly, using smaller-than-conventional RT fields or even partial tumor volume RT could be beneficial in this setting. Among host-related factors, the role of the microbiome on immunotherapy efficacy must not be overlooked nor can we neglect the role of gut irradiation in a combined RT and ICI setting. In this review we elaborate on synergistic mechanisms of immunoradiotherapy combinations, in addition to important factors to consider in future immunoradiotherapy trial designs in R/M HNSCC.
Collapse
Affiliation(s)
- Gaber Plavc
- Department of Radiation Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia; (M.O.); (P.S.)
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Tanja Jesenko
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia
| | - Miha Oražem
- Department of Radiation Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia; (M.O.); (P.S.)
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Primož Strojan
- Department of Radiation Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia; (M.O.); (P.S.)
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| |
Collapse
|
48
|
Sheftel CM, Hernandez LL. Serotonin stimulated parathyroid hormone related protein induction in the mammary epithelia by transglutaminase-dependent serotonylation. PLoS One 2020; 15:e0241192. [PMID: 33095824 PMCID: PMC7584195 DOI: 10.1371/journal.pone.0241192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 10/10/2020] [Indexed: 12/26/2022] Open
Abstract
Mammary-derived serotonin has been implicated in breast-to-bone communication during lactation by increasing parathyroid hormone related-protein (PTHrP) in the mammary gland. It is well established that PTHrP acts on the bone to liberate calcium for milk synthesis during lactation; however, the mechanism of serotonin’s regulation of PTHrP has not been fully elucidated. Recently, serotonylation has been shown to be involved in a variety of physiological processes mediated by serotonin. Therefore, we investigated whether serotonylation is involved in serotonin’s regulation of PTHrP in the mammary gland using lactogenically differentiated mouse mammary epithelial cells. We investigated the effect of increased intracellular serotonin using the antidepressant fluoxetine or 5-hydroxytryptophan (serotonin precursor), with or without transglutaminase inhibition and the corresponding action on PTHrP induction and activity. Treatment with fluoxetine or 5-hydroxytryptophan significantly increased intracellular serotonin concentrations and subsequently increased PTHrP gene expression, which was reduced with transglutaminase inhibition. Furthermore, we determined that transglutaminase activity is increased with lactogenic differentiation and 5-hydroxytryptophan or fluoxetine treatment. We investigated whether RhoA, Rac1, and Rab4 were potential serotonylation target proteins. We speculate that RhoA is potentially a serotonylation target protein. Our data suggest that serotonin regulates PTHrP induction in part through the process of serotonylation under lactogenic conditions in mouse mammary epithelial cells.
Collapse
Affiliation(s)
- Celeste M. Sheftel
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Laura L. Hernandez
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
49
|
Gupta I, Rizeq B, Vranic S, Moustafa AEA, Al Farsi H. Circulating miRNAs in HER2-Positive and Triple Negative Breast Cancers: Potential Biomarkers and Therapeutic Targets. Int J Mol Sci 2020; 21:E6750. [PMID: 32942528 PMCID: PMC7554858 DOI: 10.3390/ijms21186750] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/02/2020] [Accepted: 09/06/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is one of the most prevalent diseases among women worldwide and is highly associated with cancer-related mortality. Of the four major molecular subtypes, HER2-positive and triple-negative breast cancer (TNBC) comprise more than 30% of all breast cancers. While the HER2-positive subtype lacks estrogen and progesterone receptors and overexpresses HER2, the TNBC subtype lacks estrogen, progesterone and HER2 receptors. Although advances in molecular biology and genetics have substantially ameliorated breast cancer disease management, targeted therapies for the treatment of estrogen-receptor negative breast cancer patients are still restricted, particularly for TNBC. On the other hand, it has been demonstrated that microRNAs, miRNAs or small non-coding RNAs that regulate gene expression are involved in diverse biological processes, including carcinogenesis. Moreover, circulating miRNAs in serum/plasma are among the most promising diagnostic/therapeutic tools as they are stable and relatively easy to quantify. Various circulating miRNAs have been identified in several human cancers including specific breast cancer subtypes. This review aims to discuss the role of circulating miRNAs as potential diagnostic and prognostic biomarkers as well as therapeutic targets for estrogen-receptor negative breast cancers, HER2+ and triple negative.
Collapse
Affiliation(s)
- Ishita Gupta
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (I.G.); (B.R.); (S.V.)
- Biomedical Research Centre, Qatar University, Doha P.O. Box 2713, Qatar
| | - Balsam Rizeq
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (I.G.); (B.R.); (S.V.)
- Biomedical Research Centre, Qatar University, Doha P.O. Box 2713, Qatar
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (I.G.); (B.R.); (S.V.)
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (I.G.); (B.R.); (S.V.)
- Biomedical Research Centre, Qatar University, Doha P.O. Box 2713, Qatar
| | - Halema Al Farsi
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (I.G.); (B.R.); (S.V.)
| |
Collapse
|
50
|
Discovery of a novel selective water-soluble SMAD3 inhibitor as an antitumor agent. Bioorg Med Chem Lett 2020; 30:127396. [PMID: 32738967 DOI: 10.1016/j.bmcl.2020.127396] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/18/2022]
Abstract
Targeting the SMAD3 protein is an attractive therapeutic strategy for treating cancer, as it avoids the potential toxicities due to targeting the TGF-β signaling pathway upstream. Compound SIS3 was the first selective SMAD3 inhibitor developed that had acceptable activity, but its poor water solubility limited its development. Here, a series of SIS3 analogs was created to investigate the structure-activity relationship for inhibiting the activation of SMAD3. On the basis of this SAR, further optimization generated a water-soluble compound, 16d, which was capable of effectively blocking SMAD3 activation in vitro and had similar NK cell-mediated anticancer effects in vivo to its parent SIS3. This study not only provided a preferable lead compound, 16d, for further drug discovery or a potential tool to study SMAD3 biology, but also proved the effectiveness of our strategy for water-solubility driven optimization.
Collapse
|