51
|
Moroni S, Casettari L, Lamprou DA. 3D and 4D Printing in the Fight against Breast Cancer. BIOSENSORS 2022; 12:568. [PMID: 35892465 PMCID: PMC9394292 DOI: 10.3390/bios12080568] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
Breast cancer is the second most common cancer worldwide, characterized by a high incidence and mortality rate. Despite the advances achieved in cancer management, improvements in the quality of life of breast cancer survivors are urgent. Moreover, considering the heterogeneity that characterizes tumors and patients, focusing on individuality is fundamental. In this context, 3D printing (3DP) and 4D printing (4DP) techniques allow for a patient-centered approach. At present, 3DP applications against breast cancer are focused on three main aspects: treatment, tissue regeneration, and recovery of the physical appearance. Scaffolds, drug-loaded implants, and prosthetics have been successfully manufactured; however, some challenges must be overcome to shift to clinical practice. The introduction of the fourth dimension has led to an increase in the degree of complexity and customization possibilities. However, 4DP is still in the early stages; thus, research is needed to prove its feasibility in healthcare applications. This review article provides an overview of current approaches for breast cancer management, including standard treatments and breast reconstruction strategies. The benefits and limitations of 3DP and 4DP technologies are discussed, as well as their application in the fight against breast cancer. Future perspectives and challenges are outlined to encourage and promote AM technologies in real-world practice.
Collapse
Affiliation(s)
- Sofia Moroni
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK;
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy;
| | - Luca Casettari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy;
| | | |
Collapse
|
52
|
Breast Cancer Metastasis: Mechanisms and Therapeutic Implications. Int J Mol Sci 2022; 23:ijms23126806. [PMID: 35743249 PMCID: PMC9224686 DOI: 10.3390/ijms23126806] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the most common malignancy in women worldwide. Metastasis is the leading cause of high mortality in most cancers. Although predicting the early stage of breast cancer before metastasis can increase the survival rate, breast cancer is often discovered or diagnosed after metastasis has occurred. In general, breast cancer has a poor prognosis because it starts as a local disease and can spread to lymph nodes or distant organs, contributing to a significant impediment in breast cancer treatment. Metastatic breast cancer cells acquire aggressive characteristics from the tumor microenvironment (TME) through several mechanisms including epithelial–mesenchymal transition (EMT) and epigenetic regulation. Therefore, understanding the nature and mechanism of breast cancer metastasis can facilitate the development of targeted therapeutics focused on metastasis. This review discusses the mechanisms leading to metastasis and the current therapies to improve the early diagnosis and prognosis in patients with metastatic breast cancer.
Collapse
|
53
|
Jansson M, Lindberg J, Rask G, Svensson J, Billing O, Nazemroaya A, Berglund A, Wärnberg F, Sund M. Prognostic Value of Stromal Type IV Collagen Expression in Small Invasive Breast Cancers. Front Mol Biosci 2022; 9:904526. [PMID: 35693557 PMCID: PMC9174894 DOI: 10.3389/fmolb.2022.904526] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/09/2022] [Indexed: 11/22/2022] Open
Abstract
Breast cancer is the most common cause of cancer death among women worldwide. Localized breast cancer can be cured by surgery and adjuvant therapy, but mortality remains high for tumors that metastasize early. Type IV collagen is a basement membrane protein, and breach of this extracellular matrix structure is the first step of cancer invasion. Type IV collagen is found in the stroma of many cancers, but its role in tumor biology is unclear. Here, expression of type IV collagen in the stroma of small breast cancers was analyzed, correlated to clinically used prognostic biomarkers and patient survival. The findings were further validated in an independent gene expression data cohort. Tissue samples from 1,379 women with in situ and small invasive breast cancers (≤15 mm) diagnosed in 1986-2004 were included. Primary tumor tissue was collected into tissue microarrays. Type IV collagen expression in tissues was visualized using immunohistochemistry. Gene expression data was extracted from the Cancer Genome Atlas database. Out of 1,379 women, 856 had an invasive breast cancer and type IV collagen staining was available for 714 patients. In Kaplan-Meier analysis high type IV collagen expression was significantly associated (p = 0.026) with poorer breast cancer specific survival. There was no correlation of type IV collagen expression to clinically used prognostic biomarkers. High type IV collagen expression was clearly associated to distant metastasis (p = 0.002). In an external validation cohort (n = 1,104), high type IV collagen mRNA expression was significantly (p = 0.041) associated with poorer overall survival, with overexpression of type IV collagen mRNA in metastatic tissue. Stromal type IV collagen expression in the primary tumor correlates to poor breast cancer specific survival most likely due to a higher risk of developing distant metastasis. This ECM protein may function as biomarker to predict the risk of future metastatic disease in patients with breast cancers.
Collapse
Affiliation(s)
- Malin Jansson
- Department of Surgery and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
- *Correspondence: Malin Jansson,
| | - Jessica Lindberg
- Department of Surgery and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
| | - Gunilla Rask
- Department of Medical Biosciences/Pathology, Umeå University, Umeå, Sweden
| | - Johan Svensson
- Department of Surgery and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
- Department of Statistics, Umeå School of Business, Economics and Statistics, Umeå University, Umeå, Sweden
| | - Ola Billing
- Department of Surgery and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
| | | | - Anette Berglund
- Department of Surgery and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
| | - Fredrik Wärnberg
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Malin Sund
- Department of Surgery and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
54
|
Lee JD, Ryu WJ, Han HJ, Kim TY, Kim MH, Sohn J. Molecular Characterization of BRCA1 c.5339T>C Missense Mutation in DNA Damage Response of Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:cancers14102405. [PMID: 35626017 PMCID: PMC9139203 DOI: 10.3390/cancers14102405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/03/2022] [Accepted: 05/11/2022] [Indexed: 02/01/2023] Open
Abstract
BRCA1 L1780P BRCT domain mutation has been recognized as a pathogenic mutation in patients with breast cancer. However, the molecular significance of this mutation has not yet been studied in triple-negative breast cancer (TNBC) cells in vitro. We established MDA-MB 231, HCC1937, and HCC1395 TNBC cell lines expressing BRCA1 L1780P mutant. BRCA1 L1780P mutant TNBC cells showed increased migration and invasion capacity, as well as increased sensitivity to olaparib and carboplatin compared to BRCA1 wild-type cells. BRCA1 L1780P mutant TNBC cells showed decreased RAD51 expression and reduced nuclear RAD51 foci formation following carboplatin and olaparib treatment. The molecular interaction between p-ATM and BRCA1 was abrogated following introduction of BRCA1 L1780P mutant plasmid in TNBC cells, suggesting that the BRCA1 L1780P mutation disrupts the p-ATM-BRCA1 protein-protein interaction. We established an olaparib-resistant BRCA1 L1780P mutant TNBC cell line by chronic drug treatment. Olaparib-resistant cell lines showed upregulation of RAD51 expression upon olaparib treatment, and reduction in RAD51 expression in olaparib-resistant cells restored olaparib sensitivity. Collectively, these results suggest that the BRCA1 L1780P mutation impairs RAD51 recruitment by disrupting p-ATM-BRCA1 interaction, which is a crucial molecular factor in homologous recombination and olaparib sensitivity. Further therapeutic targeting of RAD51 in BRCA1 L1780P mutant breast cancer is warranted.
Collapse
Affiliation(s)
- Jeong Dong Lee
- Department of Human Biology and Genomics, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Won-Ji Ryu
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul 03722, Korea; (W.-J.R.); (H.J.H.); (T.Y.K.)
| | - Hyun Ju Han
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul 03722, Korea; (W.-J.R.); (H.J.H.); (T.Y.K.)
| | - Tae Yeong Kim
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul 03722, Korea; (W.-J.R.); (H.J.H.); (T.Y.K.)
| | - Min Hwan Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: (M.H.K.); (J.S.); Tel.: +82-2-2228-8135 (M.H.K. & J.S.)
| | - Joohyuk Sohn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: (M.H.K.); (J.S.); Tel.: +82-2-2228-8135 (M.H.K. & J.S.)
| |
Collapse
|
55
|
Tan K, Naylor MJ. Tumour Microenvironment-Immune Cell Interactions Influencing Breast Cancer Heterogeneity and Disease Progression. Front Oncol 2022; 12:876451. [PMID: 35646658 PMCID: PMC9138702 DOI: 10.3389/fonc.2022.876451] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/18/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is a complex, dynamic disease that acquires heterogeneity through various mechanisms, allowing cancer cells to proliferate, survive and metastasise. Heterogeneity is introduced early, through the accumulation of germline and somatic mutations which initiate cancer formation. Following initiation, heterogeneity is driven by the complex interaction between intrinsic cellular factors and the extrinsic tumour microenvironment (TME). The TME consists of tumour cells and the subsequently recruited immune cells, endothelial cells, fibroblasts, adipocytes and non-cellular components of the extracellular matrix. Current research demonstrates that stromal-immune cell interactions mediated by various TME components release environmental cues, in mechanical and chemical forms, to communicate with surrounding and distant cells. These interactions are critical in facilitating the metastatic process at both the primary and secondary site, as well as introducing greater intratumoral heterogeneity and disease complexity by exerting selective pressures on cancer cells. This can result in the adaptation of cells and a feedback loop to the cancer genome, which can promote therapeutic resistance. Thus, targeting TME and immune-stromal cell interactions has been suggested as a potential therapeutic avenue given that aspects of this process are somewhat conserved between breast cancer subtypes. This mini review will discuss emerging ideas on how the interaction of various aspects of the TME contribute to increased heterogeneity and disease progression, and the therapeutic potential of targeting the TME.
Collapse
|
56
|
Olsson LT, Williams LA, Midkiff BR, Kirk EL, Troester MA, Calhoun BC. Quantitative analysis of breast cancer tissue composition and associations with tumor subtype. Hum Pathol 2022; 123:84-92. [PMID: 35218811 PMCID: PMC9050931 DOI: 10.1016/j.humpath.2022.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment is an important determinant of breast cancer progression, but standard methods for describing the tumor microenvironment are lacking. Measures of microenvironment composition such as stromal area and immune infiltrate are labor-intensive and few large studies have systematically collected this data. However, digital histologic approaches are becoming more widely available, allowing high-throughput, quantitative estimation. We applied such methods to tissue microarrays of tumors from 1687 women (mean 4 cores per case) in the Carolina Breast Cancer Study Phase 3. Tumor composition was quantified as percentage of epithelium, stroma, adipose, and lymphocytic infiltrate (with the latter as presence/absence using a ≥1% cutoff). Composition proportions and presence/absence were evaluated in association with clinical and molecular features of breast cancer (intrinsic subtype and RNA-based risk of recurrence [ROR] scores) using multivariable linear and logistic regression. Lower stromal content was associated with aggressive tumor phenotypes, including triple-negative (31.1% vs. 41.6% in HR+/HER2-; RFD [95% CI]: -10.5%, [-13.1, -7.9]), Basal-like subtypes (29.0% vs. 44.0% in Luminal A; RFD [95% CI]: -14.9%, [-17.8, -12.0]), and high RNA-based PAM50 ROR scores (27.6% vs. 48.1% in ROR low; RFD [95% CI]: -20.5%, [24.3, 16.7]), after adjusting for age and race. HER2+ tumors also had lower stromal content, particularly among RNA-based HER2-enriched (35.2% vs. 44.0% in Luminal A; RFD [95% CI]: -8.8%, [-13.8, -3.8]). Similar associations were observed between immune infiltrate and tumor phenotypes. Quantitative digital image analysis of the breast cancer microenvironment showed significant associations with demographic characteristics and biological indicators of aggressive behavior.
Collapse
Affiliation(s)
- Linnea T Olsson
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Lindsay A Williams
- Department of Pediatrics, University of Minnesota School of Medicine, Minneapolis, MN, 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Bentley R Midkiff
- Pathology Services Core, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Erin L Kirk
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Melissa A Troester
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Benjamin C Calhoun
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA; Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
57
|
Putri SD, Nanza SRY, Widodo I, Purnomosari D. The Association of Intra-Tumoral and Stromal Vitamin D Receptor (VDR) Expressions with Molecular Subtypes and Clinicopathological Factors in Breast Carcinoma. Asian Pac J Cancer Prev 2022; 23:1169-1175. [PMID: 35485672 PMCID: PMC9375625 DOI: 10.31557/apjcp.2022.23.4.1169] [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: 02/05/2021] [Accepted: 04/08/2022] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE This study aimed to investigate the association between intra-tumoral and stromal VDR expressions with molecular subtypes and clinicopathological factors. METHODS A total of 75 formalin-fixed paraffin embedded tissue samples were stained using immunohistochemical methods. The VDR expressions were measured by counting brown-stained nuclei in intra-tumoral and stromal areas. The association of VDR expression with molecular subtypes and clinopathological factors was examined. Statistical analysis was performed by chi square tests. RESULTS High intra-tumoral VDR expression was found in carcinomas with luminal molecular subtypes (p=0.039) and low histological degrees (p=0.035). High VDR expression in the stroma was found in breast carcinomas with large tumor sizes. CONCLUSIONS High intra-tumoral VDR expression is found in breast carcinomas with luminal subtypes and low histological grade (I/II). Both factors are known to have a good prognosis. These findings further strengthen the function of VDR as anti-tumorigenesis.
Collapse
Affiliation(s)
- Sukma Diani Putri
- Postgraduate Program of Biomedical Sciences, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Indonesia.
- Indramayu State Polytechnic.
| | - Siti Rahma Yunianda Nanza
- Postgraduate Program of Biomedical Sciences, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Indonesia.
| | - Irianiwati Widodo
- Department of Anatomic Pathology, Faculty of Medicine, Public Health and Nursing Universitas Gadjah Mada, Indonesia.
| | - Dewajani Purnomosari
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Indonesia.
| |
Collapse
|
58
|
Radharani NNV, Yadav AS, Nimma R, Kumar TVS, Bulbule A, Chanukuppa V, Kumar D, Patnaik S, Rapole S, Kundu GC. Tumor-associated macrophage derived IL-6 enriches cancer stem cell population and promotes breast tumor progression via Stat-3 pathway. Cancer Cell Int 2022; 22:122. [PMID: 35300689 PMCID: PMC8932105 DOI: 10.1186/s12935-022-02527-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/21/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) play crucial role in tumor progression, drug resistance and relapse in various cancers. CSC niche is comprised of various stromal cell types including Tumor-associated macrophages (TAMs). Extrinsic ques derived from these cells help in maintenance of CSC phenotype. TAMs have versatile roles in tumor progression however their function in enrichment of CSC is poorly explored. METHODS Mouse macrophages (RAW264.7) cells were activated by interaction with conditioned media (CM) of murine breast cancer cells (4T1) into TAMs and the effect of activated macrophage (TAM) derived factors was examined on enrichment of cancer stem cells (CSCs) and tumor growth using in vitro and in vivo models. RESULTS In this study, we report that macrophages upon interaction with breast cancer cells activate tumor promoting function and exhibit differential expression of various proteins as shown by secretome analysis using proteomics studies. Based on secretome data, we found that Interleukin-6 (IL-6) is one of the up-regulated genes expressed in activated macrophages. Further, we confirm that TAMs produce high levels of IL-6 and breast cancer cell derived factors induce IL-6 production in activated macrophages via p38-MAPK pathway. Furthermore, we demonstrate that tumor activated macrophages induce enrichment of CSCs and expression of CSC specific transcription factors such as Sox-2, Oct-3/4 and Nanog in breast cancer cells. We further prove that TAM derived IL-6 plays a key role in TAM mediated CSC enrichment through activation of Signal transducer and activator of transcription 3 (STAT-3) signaling. TAM derived IL-6 influences breast cancer cell migration and angiogenesis. Moreover, our in vivo findings indicated that TAM derived IL-6 induces CSC population and resulting tumor growth in breast cancer. CONCLUSION These finding provide evidence that TAM derived IL-6 plays a major role in CSC enrichment and tumor progression in breast cancer and IL-6 and its regulated signalling network may act as potential therapeutic target for management of breast cancer.
Collapse
Affiliation(s)
- N N V Radharani
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, 411007, India.,School of Biotechnology, KIIT Deemed To Be University, Bhubaneswar, 751 024, India
| | - Amit S Yadav
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, 411007, India.,School of Biotechnology, KIIT Deemed To Be University, Bhubaneswar, 751 024, India
| | - Ramakrishna Nimma
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, 411007, India
| | - T V Santosh Kumar
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, 411007, India
| | - Anuradha Bulbule
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, 411007, India
| | - Venkatesh Chanukuppa
- Proteomics Laboratory, National Centre for Cell Science (NCCS), Pune, 411007, India
| | - Dhiraj Kumar
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Srinivas Patnaik
- School of Biotechnology, KIIT Deemed To Be University, Bhubaneswar, 751 024, India
| | - Srikanth Rapole
- Proteomics Laboratory, National Centre for Cell Science (NCCS), Pune, 411007, India
| | - Gopal C Kundu
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, 411007, India. .,School of Biotechnology, KIIT Deemed To Be University, Bhubaneswar, 751 024, India. .,Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed To Be University, Bhubaneswar, 751024, India.
| |
Collapse
|
59
|
Xu C, Wang F, Hao L, Liu J, Shan B, Lv S, Han X, Pan Y, Niu Y. Expression Patterns of Ezrin and AJAP1 and Clinical Significance in Breast Cancer. Front Oncol 2022; 12:831507. [PMID: 35311087 PMCID: PMC8931223 DOI: 10.3389/fonc.2022.831507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/02/2022] [Indexed: 12/29/2022] Open
Abstract
Ezrin and adherens junction-associated protein 1 (AJAP1) are structural proteins which are involved in numerous human malignancies. However, little is known about the relationship between them in breast cancer. This study was set out to investigate the relationship between them and to further explore the mechanism of AJAP1-mediating cytoskeleton in breast cancer progression. Ezrin and AJAP1 expressions were detected in 377 samples of breast cancer by immunohistochemistry, and different expression patterns between AJAP1 and Ezrin with clinicopathological parameters were analyzed. Besides, univariate and multivariate Cox models were used to evaluate their prognostic potential. Enzyme-linked immunosorbent assay, Western blot, qRT-PCR, and phalloidin staining of F-actin were used to explore the relationship and the mechanism between AJAP1 and Ezrin in cytoskeleton arrangement. 377 cases of breast cancer results showed that AJAP1 expression was negatively related with histological grade and lymph node involvement and could be an independent prognosis marker of breast cancer. AJAP1 expression tended to be higher in the Ezrin-negative expression case. Patients with AJAP1negative and Ezrinpositive expression had a worse prognosis (p < 0.0001) and shorter DFS (p = 0.015). More importantly, AJAP1 depletion increased the cell ability of F-actin formation through promoting Ezrin expression. AJAP1 depletion might mediate breast cancer malignancy potential through promoting Ezrin expression and cytoskeleton formation.
Collapse
Affiliation(s)
- Cong Xu
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Feng Wang
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Li Hao
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jing Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuhua Lv
- Department of Pathology, Tianjin Union Medical Center, Tianjin People’s Hospital, Tianjin, China
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Xinghua Han, ; Yueyin Pan, ; Yun Niu,
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Xinghua Han, ; Yueyin Pan, ; Yun Niu,
| | - Yun Niu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- *Correspondence: Xinghua Han, ; Yueyin Pan, ; Yun Niu,
| |
Collapse
|
60
|
Silva D, Quintas C, Gonçalves J, Fresco P. Contribution of adrenergic mechanisms for the stress-induced breast cancer carcinogenesis. J Cell Physiol 2022; 237:2107-2127. [PMID: 35243626 DOI: 10.1002/jcp.30707] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/21/2022] [Accepted: 02/12/2022] [Indexed: 12/11/2022]
Abstract
Breast cancer is the most common and deadliest type of cancer in women. Stress exposure has been associated with carcinogenesis and the stress released neurotransmitters, noradrenaline and adrenaline, and their cognate receptors, can participate in the carcinogenesis process, either by regulating tumor microenvironment or by promoting systemic changes. This work intends to provide an overview of the research done in this area and try to unravel the role of adrenergic ligands in the context of breast carcinogenesis. In the initiation phase, adrenergic signaling may favor neoplastic transformation of breast epithelial cells whereas, during cancer progression, may favor the metastatic potential of breast cancer cells. Additionally, adrenergic signaling can alter the function and activity of other cells present in the tumor microenvironment towards a protumor phenotype, namely macrophages, fibroblasts, and by altering adipocyte's function. Adrenergic signaling also promotes angiogenesis and lymphangiogenesis and, systemically, may induce the formation of preneoplastic niches, cancer-associated cachexia and alterations in the immune system which contribute for the loss of quality of life of breast cancer patients and their capacity to fight cancer. Most studies points to a major contribution of β2 -adrenoceptor activated pathways on these effects. The current knowledge of the mechanistic pathways activated by β2 -adrenoceptors in physiology and pathophysiology, the availability of selective drugs approved for clinical use and a deeper knowledge of the basic cellular and molecular pathways by which adrenergic stimulation may influence cancer initiation and progression, opens the possibility to use new therapeutic alternatives to improve efficacy of breast cancer treatments.
Collapse
Affiliation(s)
- Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Clara Quintas
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
61
|
Song T, Markham KK, Li Z, Muller KE, Greenham K, Kuang R. Detecting spatially co-expressed gene clusters with functional coherence by graph-regularized convolutional neural network. Bioinformatics 2022; 38:1344-1352. [PMID: 34864909 DOI: 10.1093/bioinformatics/btab812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/29/2021] [Accepted: 11/29/2021] [Indexed: 01/05/2023] Open
Abstract
MOTIVATION Clustering spatial-resolved gene expression is an essential analysis to reveal gene activities in the underlying morphological context by their functional roles. However, conventional clustering analysis does not consider gene expression co-localizations in tissue for detecting spatial expression patterns or functional relationships among the genes for biological interpretation in the spatial context. In this article, we present a convolutional neural network (CNN) regularized by the graph of protein-protein interaction (PPI) network to cluster spatially resolved gene expression. This method improves the coherence of spatial patterns and provides biological interpretation of the gene clusters in the spatial context by exploiting the spatial localization by convolution and gene functional relationships by graph-Laplacian regularization. RESULTS In this study, we tested clustering the spatially variable genes or all expressed genes in the transcriptome in 22 Visium spatial transcriptomics datasets of different tissue sections publicly available from 10× Genomics and spatialLIBD. The results demonstrate that the PPI-regularized CNN constantly detects gene clusters with coherent spatial patterns and significantly enriched by gene functions with the state-of-the-art performance. Additional case studies on mouse kidney tissue and human breast cancer tissue suggest that the PPI-regularized CNN also detects spatially co-expressed genes to define the corresponding morphological context in the tissue with valuable insights. AVAILABILITY AND IMPLEMENTATION Source code is available at https://github.com/kuanglab/CNN-PReg. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Tianci Song
- Department of Computer Science and Engineering, University of Minnesota Twin Cities, Minneapolis, MN 55414, USA
| | - Kathleen K Markham
- Department of Plant and Microbial Biology, University of Minnesota Twin Cities, Minneapolis, MN 55414, USA
| | - Zhuliu Li
- Department of Computer Science and Engineering, University of Minnesota Twin Cities, Minneapolis, MN 55414, USA
| | - Kristen E Muller
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Kathleen Greenham
- Department of Plant and Microbial Biology, University of Minnesota Twin Cities, Minneapolis, MN 55414, USA
| | - Rui Kuang
- Department of Computer Science and Engineering, University of Minnesota Twin Cities, Minneapolis, MN 55414, USA
| |
Collapse
|
62
|
Beeghly GF, Amofa KY, Fischbach C, Kumar S. Regulation of Tumor Invasion by the Physical Microenvironment: Lessons from Breast and Brain Cancer. Annu Rev Biomed Eng 2022; 24:29-59. [PMID: 35119915 DOI: 10.1146/annurev-bioeng-110220-115419] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The success of anticancer therapies is often limited by heterogeneity within and between tumors. While much attention has been devoted to understanding the intrinsic molecular diversity of tumor cells, the surrounding tissue microenvironment is also highly complex and coevolves with tumor cells to drive clinical outcomes. Here, we propose that diverse types of solid tumors share common physical motifs that change in time and space, serving as universal regulators of malignancy. We use breast cancer and glioblastoma as instructive examples and highlight how invasion in both diseases is driven by the appropriation of structural guidance cues, contact-dependent heterotypic interactions with stromal cells, and elevated interstitial fluid pressure and flow. We discuss how engineering strategies show increasing value for measuring and modeling these physical properties for mechanistic studies. Moreover, engineered systems offer great promise for developing and testing novel therapies that improve patient prognosis by normalizing the physical tumor microenvironment. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 24 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Garrett F Beeghly
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA;
| | - Kwasi Y Amofa
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, USA; .,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA; .,Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, USA
| | - Sanjay Kumar
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, USA; .,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, USA.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
63
|
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
|
64
|
Bakshi HA, Faruck HL, Ravesh Z, Ansari P, Hannan JMA, Hashimoto R, Takayama K, Farzand R, Nasef MM, Mensah A, Aljabali AAA, Mishra V, Charbe NB, Goyal R, Negi P, Serrano-Aroca Á, Bahar B, El-Tanani M, Courtenay AJ, McCarron P, Jack IG, Tambuwala MM. Therapeutic Potential of Cannabinoids on Tumor Microenvironment: A Molecular Switch in Neoplasia Transformation. Integr Cancer Ther 2022; 21:15347354221096766. [PMID: 35796303 PMCID: PMC9425895 DOI: 10.1177/15347354221096766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/02/2022] [Accepted: 04/09/2022] [Indexed: 11/24/2022] Open
Abstract
The efficacy of chemotherapy depends on the tumor microenvironment. This microenvironment consists of a complex cellular network that can exert both stimulatory and inhibitory effects on tumor genesis. Given the increasing interest in the effectiveness of cannabis, cannabinoids have gained much attention as a potential chemotherapy drug. Cannabinoids are a group of marker compounds found in Cannabis sativa L., more commonly known as marijuana, a psychoactive drug used since ancient times for pain management. Although the anticancer potential of C. sativa, has been recognized previously, increased attention was generated after discovering the endocannabinoid system and the successful production of cannabinoid receptors. In vitro and in vivo studies on various tumor models have shown therapeutic efficiency by modifying the tumor microenvironment. However, despite extensive attention regarding potential therapeutic implications of cannabinoids, considerable clinical and preclinical analysis is needed to adequately define the physiological, pharmacological, and medicinal aspects of this range of compounds in various disorders covered in this review. This review summarizes the key literature surrounding the role of cannabinoids in the tumor microenvironment and their future promise in cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rabia Farzand
- University of Huddersfield, Queensgate, Huddersfield, UK
| | | | | | | | - Vijay Mishra
- Lovely Professional University, Phagwara, Punjab, India
| | | | - Rohit Goyal
- Shoolini University, Solan, Himachal Pradesh, India
| | - Poonam Negi
- Shoolini University, Solan, Himachal Pradesh, India
| | | | | | | | | | - Paul McCarron
- Ulster University, Coleraine, County Londonderry, UK
| | - Iain G. Jack
- Ulster University, Coleraine, County Londonderry, UK
| | | |
Collapse
|
65
|
Apple polyphenol phloretin complexed with ruthenium is capable of reprogramming the breast cancer microenvironment through modulation of PI3K/Akt/mTOR/VEGF pathways. Toxicol Appl Pharmacol 2022; 434:115822. [PMID: 34896434 DOI: 10.1016/j.taap.2021.115822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/10/2021] [Accepted: 12/05/2021] [Indexed: 12/15/2022]
Abstract
Our recent investigation directed to synthesize a novel ruthenium-phloretin complex accompanied by the study of antioxidant in addition to DNA binding capabilities, to determine the chemotherapeutic activity against breast carcinoma in vitro and in vivo. Ruthenium-phloretin complex was synthesized and characterized by different spectroscopic methods. The complex was further investigated to determine its efficacy in both MCF-7 and MDA-MB-231 human carcinoma cell lines and finally in an in vivo model of mammary carcinogenesis induced by DMBA in rats. Our studies confirm that the chelation of the metal and ligand was materialize by the 3-OH and 9-OH functional groups of the ligand and the complex is found crystalline and was capable of intercalating with CT-DNA. The complex was capable of reducing cellular propagation and initiate apoptotic events in MCF-7 and MDA-MB-231 breast carcinoma cell lines. Ruthenium-phloretin complex could modulate p53 intervene apoptosis in the breast carcinoma, initiated by the trail of intrinsic apoptosis facilitated through Bcl2 and Bax and at the same time down regulating the PI3K/Akt/mTOR pathway coupled with MMP9 regulated tumor invasive pathways. Ruthenium-phloretin chemotherapy could interrupt, revoke or suspend the succession of breast carcinoma by altering intrinsic apoptosis along with the anti-angiogenic pathway.
Collapse
|
66
|
Krishnan R, Patel PS, Hakem R. BRCA1 and Metastasis: Outcome of Defective DNA Repair. Cancers (Basel) 2021; 14:cancers14010108. [PMID: 35008272 PMCID: PMC8749860 DOI: 10.3390/cancers14010108] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary BRCA1 has critical functions in accurately repairing double stand breaks in the DNA through a process known as homologous recombination. BRCA1 also has various functions in other cellular processes that safeguard the genome. Thus, mutations or silencing of this tumor suppressor significantly increases the risk of developing breast, ovarian, and other cancers. Metastasis refers to the spread of cancer to other parts of the body and is the leading cause of cancer-related deaths. In this review, we discuss the mechanisms by which BRCA1 mutations contribute to the metastatic and aggressive nature of the tumor cells. Abstract Heritable mutations in BRCA1 and BRCA2 genes are a major risk factor for breast and ovarian cancer. Inherited mutations in BRCA1 increase the risk of developing breast cancers by up to 72% and ovarian cancers by up to 69%, when compared to individuals with wild-type BRCA1. BRCA1 and BRCA2 (BRCA1/2) are both important for homologous recombination-mediated DNA repair. The link between BRCA1/2 mutations and high susceptibility to breast cancer is well established. However, the potential impact of BRCA1 mutation on the individual cell populations within a tumor microenvironment, and its relation to increased aggressiveness of cancer is not well understood. The objective of this review is to provide significant insights into the mechanisms by which BRCA1 mutations contribute to the metastatic and aggressive nature of the tumor cells.
Collapse
Affiliation(s)
- Rehna Krishnan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; (R.K.); (P.S.P.)
| | - Parasvi S. Patel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; (R.K.); (P.S.P.)
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Razqallah Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; (R.K.); (P.S.P.)
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence: or
| |
Collapse
|
67
|
Current Advancements of Plant-Derived Agents for Triple-Negative Breast Cancer Therapy through Deregulating Cancer Cell Functions and Reprogramming Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms222413571. [PMID: 34948368 PMCID: PMC8703661 DOI: 10.3390/ijms222413571] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is defined based on the absence of estrogen, progesterone, and human epidermal growth factor receptor 2 receptors. Currently, chemotherapy is the major therapeutic approach for TNBC patients; however, poor prognosis after a standard chemotherapy regimen is still commonplace due to drug resistance. Abnormal tumor metabolism and infiltrated immune or stromal cells in the tumor microenvironment (TME) may orchestrate mammary tumor growth and metastasis or give rise to new subsets of cancer cells resistant to drug treatment. The immunosuppressive mechanisms established in the TME make cancer cell clones invulnerable to immune recognition and killing, and turn immune cells into tumor-supporting cells, hence allowing cancer growth and dissemination. Phytochemicals with the potential to change the tumor metabolism or reprogram the TME may provide opportunities to suppress cancer metastasis and/or overcome chemoresistance. Furthermore, phytochemical intervention that reprograms the TME away from favoring immunoevasion and instead towards immunosurveillance may prevent TNBC metastasis and help improve the efficacy of combination therapies as phyto-adjuvants to combat drug-resistant TNBC. In this review, we summarize current findings on selected bioactive plant-derived natural products in preclinical mouse models and/or clinical trials with focus on their immunomodulatory mechanisms in the TME and their roles in regulating tumor metabolism for TNBC prevention or therapy.
Collapse
|
68
|
Osteocytic Connexin Hemichannels Modulate Oxidative Bone Microenvironment and Breast Cancer Growth. Cancers (Basel) 2021; 13:cancers13246343. [PMID: 34944962 PMCID: PMC8699531 DOI: 10.3390/cancers13246343] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022] Open
Abstract
Osteocytes, the most abundant bone cell types embedded in the mineral matrix, express connexin 43 (Cx43) hemichannels that play important roles in bone remodeling and osteocyte survival. Estrogen deficiency decreases osteocytic Cx43 hemichannel activity and causes a loss in osteocytes' resistance to oxidative stress (OS). In this study, we showed that OS reduced the growth of both human (MDA-MB-231) and murine (Py8119) breast cancer cells. However, co-culturing these cells with osteocytes reduced the inhibitory effect of OS on breast cancer cells, and this effect was ablated by the inhibition of Cx43 hemichannels. Py8119 cells were intratibially implanted in the bone marrow of ovariectomized (OVX) mice to determine the role of osteocytic Cx43 hemichannels in breast cancer bone metastasis in response to OS. Two transgenic mice overexpressing dominant-negative Cx43 mutants, R76W and Δ130-136, were adopted for this study; the former inhibits gap junctions while the latter inhibits gap junctions and hemichannels. Under normal conditions, Δ130-136 mice had significantly more tumor growth in bone than that in WT and R76W mice. OVX increased tumor growth in R76W but had no significant effect on WT mice. In contrast, OVX reduced tumor growth in Δ130-136 mice. To confirm the role of OS, WT and Δ130-136 mice were administered the antioxidant N-acetyl cysteine (NAC). NAC increased tumor burden and growth in Δ130-136 mice but not in WT mice. Together, the data suggest that osteocytes and Cx43 hemichannels play pivotal roles in modulating the oxidative microenvironment and breast cancer growth in the bone.
Collapse
|
69
|
Rohini M, Vairamani M, Selvamurugan N. TGF-β1-stimulation of NFATC2 and ATF3 proteins and their interaction for matrix metalloproteinase 13 expression in human breast cancer cells. Int J Biol Macromol 2021; 192:1325-1330. [PMID: 34687766 DOI: 10.1016/j.ijbiomac.2021.10.099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/30/2021] [Accepted: 10/14/2021] [Indexed: 01/25/2023]
Abstract
Activating transcription factor 3 (ATF3), an inducible stress gene, is stimulated by transforming growth factor-beta1 (TGF-β1) in a protracted and relentless manner in human mammary cancer cells (hBC cells; MDA-MB231). The molecular mechanism behind this stable expression of ATF3 via TGF-β1 in MDA-MB231 cells is unknown. This study found that TGF-β1 stimulated the expression of the nuclear factor of activated T Cells 2 (NFATC2) in MDA-MB231 cells and provided evidence of its interaction with ATF3. The functional characterization of NFATC2 in association with ATF3 was determined by silencing of NFATC2 using siRNA. Knock-down of NFATC2 decreased the expression of both ATF3 and its target gene MMP13 (matrix metalloproteinase 13, a critical invasive gene) in hBC cells. Chromatin immunoprecipitation revealed that TGF-β1 promoted NFATC2 binding and NFATC2-ATF3 complex binding at the MMP13 promoter region, whereas silencing of NFATC2 decreased their binding in hBC cells. Thus, we uncovered the mechanism of interaction between NFATC2 and ATF3 regulated by TGF-β1, and NFATC2 acted as a pivotal factor in providing ATF3 stability and further drove MMP13 transcription. Targeting NFATC2 and blocking its association with ATF3 could therefore help to slow the progression of breast cancer.
Collapse
Affiliation(s)
- M Rohini
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - M Vairamani
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
70
|
Abd El-Aziz YS, Gillson J, Jansson PJ, Sahni S. Autophagy: A promising target for triple negative breast cancers. Pharmacol Res 2021; 175:106006. [PMID: 34843961 DOI: 10.1016/j.phrs.2021.106006] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 01/18/2023]
Abstract
Triple negative breast cancer (TNBC) is the most aggressive type of breast cancers which constitutes about 15% of all breast cancer cases and characterized by negative expression of hormonal receptors and human epidermal growth factor receptor 2 (HER2). Thus, endocrine and HER2 targeted therapies are not effective toward TNBCs, and they mainly rely on chemotherapy and surgery for treatment. Despite recent advances in chemotherapy, 40% of TNBC patients develop a metastatic relapse and recurrence. Therefore, understanding the molecular profile of TNBC is warranted to identify targets that can be selected for the development of a new and effective therapeutic approach. Autophagy is an internal defensive mechanism that allows the cells to survive under different stressors. It has been well known that autophagy exerts a crucial role in cancer progression. The critical role of autophagy in TNBC progression is emerging in recent years. This review will discuss autophagic pathway, how autophagy affects TNBC progression and recent therapeutic approaches that can target autophagy as a new treatment modality.
Collapse
Affiliation(s)
- Yomna S Abd El-Aziz
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Kolling Institute of Medical Research, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Josef Gillson
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Kolling Institute of Medical Research, St Leonards, NSW, Australia
| | - Patric J Jansson
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Kolling Institute of Medical Research, St Leonards, NSW, Australia; Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Sumit Sahni
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Kolling Institute of Medical Research, St Leonards, NSW, Australia.
| |
Collapse
|
71
|
Giorello MB, Matas A, Marenco P, Davies KM, Borzone FR, Calcagno MDL, García-Rivello H, Wernicke A, Martinez LM, Labovsky V, Chasseing NA. CD1a- and CD83-positive dendritic cells as prognostic markers of metastasis development in early breast cancer patients. Breast Cancer 2021; 28:1328-1339. [PMID: 34240315 DOI: 10.1007/s12282-021-01270-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Dendritic cells (DCs) are the most potent antigen-presenting cells that play a major role in initiating the antitumor immune response in different types of cancer. However, the prognostic significance of the accumulation of these cells in human early breast tumors is not totally clear. The aim of this study is to evaluate the prognostic relevance of CD1a( +) and CD83( +) dendritic cells in early breast cancer patients. METHODS We conducted immunohistochemical assays to determine the number of stromal CD1a( +) and CD83( +) DCs in primary tumors from early invasive ductal breast cancer patients, and analyzed their association with clinico-pathological characteristics. RESULTS Patients with high CD1a( +) DC number had lower risk of bone metastatic occurrence, as well as, longer disease-free survival (DFS), bone metastasis-free survival (BMFS) and overall survival (OS). Moreover, CD1a( +) DC number was an independent prognostic factor for BMFS and OS. In contrast, we found that patients with high number of CD83( +) DCs had lower risk of mix (bone and visceral)-metastatic occurrence. Likewise, these patients presented better prognosis with longer DFS, mix-MFS and OS. Furthermore, CD83( +) DC number was an independent prognostic factor for DFS and OS. CONCLUSION The quantification of the stromal infiltration of DCs expressing CD1a or CD83 in early invasive breast cancer patients serves to indicate the prognostic risk of developing metastasis in a specific site.
Collapse
Affiliation(s)
- María Belén Giorello
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Instituto de Biología Y Medicina Experimental, Vuelta de Obligado 2490, Ciudad Autónoma de Buenos Aires, CP 1428, Buenos Aires, Argentina
| | - Ayelén Matas
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Instituto de Biología Y Medicina Experimental, Vuelta de Obligado 2490, Ciudad Autónoma de Buenos Aires, CP 1428, Buenos Aires, Argentina
| | - Pablo Marenco
- Departamento de Anatomía Patológica, Hospital Italiano, Juan Domingo Perón 4190, Ciudad Autónoma de Buenos Aires, CP 1181, Buenos Aires, Argentina
| | - Kevin Mauro Davies
- Departamento de Anatomía Patológica, Hospital Italiano, Juan Domingo Perón 4190, Ciudad Autónoma de Buenos Aires, CP 1181, Buenos Aires, Argentina
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Instituto de Biología Y Medicina Experimental, Vuelta de Obligado 2490, Ciudad Autónoma de Buenos Aires, CP 1428, Buenos Aires, Argentina
| | - María de Luján Calcagno
- Facultad de Farmacia Y Bioquímica, Universidad de Buenos Aires, Junín 954, Ciudad Autónoma de Buenos Aires, CP 1113, Buenos Aires, Argentina
| | - Hernán García-Rivello
- Departamento de Anatomía Patológica, Hospital Italiano, Juan Domingo Perón 4190, Ciudad Autónoma de Buenos Aires, CP 1181, Buenos Aires, Argentina
| | - Alejandra Wernicke
- Departamento de Anatomía Patológica, Hospital Italiano, Juan Domingo Perón 4190, Ciudad Autónoma de Buenos Aires, CP 1181, Buenos Aires, Argentina
| | - Leandro Marcelo Martinez
- Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Vivian Labovsky
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Instituto de Biología Y Medicina Experimental, Vuelta de Obligado 2490, Ciudad Autónoma de Buenos Aires, CP 1428, Buenos Aires, Argentina.
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Instituto de Biología Y Medicina Experimental, Vuelta de Obligado 2490, Ciudad Autónoma de Buenos Aires, CP 1428, Buenos Aires, Argentina.
| |
Collapse
|
72
|
Alraouji NN, Hendrayani SF, Ghebeh H, Al-Mohanna FH, Aboussekhra A. Osteoprotegerin (OPG) mediates the anti-carcinogenic effects of normal breast fibroblasts and targets cancer stem cells through inhibition of the β-catenin pathway. Cancer Lett 2021; 520:374-384. [PMID: 34416336 DOI: 10.1016/j.canlet.2021.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 01/20/2023]
Abstract
Normal breast fibroblasts (NBFs) support and maintain the architecture of the organ, and can also suppress tumorigenesis. However, the mechanisms involved are not fully understood. We have shown here that NBFs suppress breast carcinogenesis through secretion of osteoprotegerin (OPG), a soluble decoy receptor for the Receptor Activator of NF-κB ligand (RANKL). Indeed, NBFs and human recombinant OPG (rOPG), suppressed breast cancer cells proliferation and motility through inhibition of the epithelial-to-mesenchymal transition (EMT) process both in vitro and in vivo. Additionally, rOPG inhibited the IL-6/STAT3 and NF-κB pathways as well as the OPG gene, which turned out to be STAT3-regulated. This was confirmed using denosumab, a RANKL-targeted antibody, which also inhibited NF-κB, down-regulated OPG and repressed EMT in breast cancer cells grown in 2D and 3D. Importantly, both rOPG and denosumab targeted cancer stem cells (CSCs). This was mediated through inhibition of the CSC-related pathway β-catenin. Moreover, rOPG reduced tumor growth and inhibited breast CSC biomarkers in orthotopic humanized breast tumors. Therefore, normal mammary fibroblasts can suppress carcinogenesis through OPG, which constitutes great potential as preventive and/or therapeutic molecule for breast carcinomas.
Collapse
Affiliation(s)
- Noura N Alraouji
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Siti-Fauziah Hendrayani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Hazem Ghebeh
- Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, MBC#03, Riyadh, 11211, Saudi Arabia
| | - Falah H Al-Mohanna
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Abdelilah Aboussekhra
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia.
| |
Collapse
|
73
|
Mohammad Mirzaei N, Su S, Sofia D, Hegarty M, Abdel-Rahman MH, Asadpoure A, Cebulla CM, Chang YH, Hao W, Jackson PR, Lee AV, Stover DG, Tatarova Z, Zervantonakis IK, Shahriyari L. A Mathematical Model of Breast Tumor Progression Based on Immune Infiltration. J Pers Med 2021; 11:jpm11101031. [PMID: 34683171 PMCID: PMC8540934 DOI: 10.3390/jpm11101031] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 01/03/2023] Open
Abstract
Breast cancer is the most prominent type of cancer among women. Understanding the microenvironment of breast cancer and the interactions between cells and cytokines will lead to better treatment approaches for patients. In this study, we developed a data-driven mathematical model to investigate the dynamics of key cells and cytokines involved in breast cancer development. We used gene expression profiles of tumors to estimate the relative abundance of each immune cell and group patients based on their immune patterns. Dynamical results show the complex interplay between cells and molecules, and sensitivity analysis emphasizes the direct effects of macrophages and adipocytes on cancer cell growth. In addition, we observed the dual effect of IFN-γ on cancer proliferation, either through direct inhibition of cancer cells or by increasing the cytotoxicity of CD8+ T-cells.
Collapse
Affiliation(s)
- Navid Mohammad Mirzaei
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (N.M.M.); (S.S.); (D.S.); (M.H.)
| | - Sumeyye Su
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (N.M.M.); (S.S.); (D.S.); (M.H.)
| | - Dilruba Sofia
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (N.M.M.); (S.S.); (D.S.); (M.H.)
| | - Maura Hegarty
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (N.M.M.); (S.S.); (D.S.); (M.H.)
| | - Mohamed H. Abdel-Rahman
- Department of Ophthalmology, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (M.H.A.-R.); (C.M.C.); (D.G.S.)
| | - Alireza Asadpoure
- Department of Civil and Environmental Engineering, University of Massachusetts, Dartmouth, MA 02747, USA;
| | - Colleen M. Cebulla
- Department of Ophthalmology, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (M.H.A.-R.); (C.M.C.); (D.G.S.)
| | - Young Hwan Chang
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, OR 97239, USA; (Y.H.C.); (Z.T.)
| | - Wenrui Hao
- Department of Mathematics, The Pennsylvania State University, University Park, PA 16802, USA;
| | - Pamela R. Jackson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic Arizona, Phoenix, AZ 85054, USA;
| | - Adrian V. Lee
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15219, USA;
| | - Daniel G. Stover
- Department of Ophthalmology, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (M.H.A.-R.); (C.M.C.); (D.G.S.)
| | - Zuzana Tatarova
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, OR 97239, USA; (Y.H.C.); (Z.T.)
| | - Ioannis K. Zervantonakis
- Department of Bioengineering, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15219, USA;
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (N.M.M.); (S.S.); (D.S.); (M.H.)
- Correspondence:
| |
Collapse
|
74
|
Fujikawa T, Sanada F, Taniyama Y, Shibata K, Katsuragi N, Koibuchi N, Akazawa K, Kanemoto Y, Kuroyanagi H, Shimazu K, Rakugi H, Morishita R. Periostin Exon-21 Antibody Neutralization of Triple-Negative Breast Cancer Cell-Derived Periostin Regulates Tumor-Associated Macrophage Polarization and Angiogenesis. Cancers (Basel) 2021; 13:cancers13205072. [PMID: 34680221 PMCID: PMC8533925 DOI: 10.3390/cancers13205072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Despite remarkable advances in breast cancer treatment, few strategies other than standard cytotoxic chemotherapy are available for patients with triple-negative breast cancer (TNBC) due to the lack of therapeutic target molecules. TNBC is still the most aggressive subtype, with a high risk of recurrence and metastasis within 2 years after initial treatment. Thus, there is an unmet medical need to develop new treatments for metastatic and recurrent TNBC patients. In this study we tested a new antibody, targeting extracellular periostin protein alternative splicing variants, which are induced by conventional chemotherapy or during the process of endothelial mesenchymal transition. This antibody reduced periostin-secreting TNBC in a mouse xenograft model, accompanied by a decrease in the number of M2 tumor-associated macrophages and tumor vessels. Periostin alternative splicing variants might be a specific and safe therapeutic target in patients with TNBC. Abstract Periostin (Pn) is involved in multiple processes of cancer progression. Previously, we reported that Pn expression is correlated with mesenchymal tumor markers and poor prognosis in triple-negative breast cancer (TNBC). In the TNBC xenograft model, chemotherapy increased expression of a Pn alternative splicing variant (ASV) with exon 21, and administration of the neutralizing antibody against Pn with exon 21 (Pn-21 Ab) overcame chemoresistance with a reduction in the mesenchymal cancer cell fraction. In the present study, the role of Pn ASV with exon 21 in TNBC progression has been addressed. We first established a stable cell line carrying a fluorescence-based splicing reporter. Pn-positive TNBC has higher expression of genes related to tumor-associated macrophage (TAM) recruitment and ECM-receptor interaction than Pn-negative cells. In a xenograft model, only Pn-positive cells initiated tumor formation, and the Pn-21 Ab suppressed tumor cell growth, accompanied by decreased M2 TAM polarization and the number of tumor vessels. These data suggest that cancer cell-derived Pn ASV educates TAMs and regulates angiogenesis, which in turn establishes a microenvironmental niche that is supportive of TNBC.
Collapse
Affiliation(s)
- Tatsuya Fujikawa
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; (T.F.); (F.S.); (Y.T.); (K.S.); (N.K.); (N.K.)
| | - Fumihiro Sanada
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; (T.F.); (F.S.); (Y.T.); (K.S.); (N.K.); (N.K.)
| | - Yoshiaki Taniyama
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; (T.F.); (F.S.); (Y.T.); (K.S.); (N.K.); (N.K.)
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan;
| | - Kana Shibata
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; (T.F.); (F.S.); (Y.T.); (K.S.); (N.K.); (N.K.)
| | - Naruto Katsuragi
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; (T.F.); (F.S.); (Y.T.); (K.S.); (N.K.); (N.K.)
| | - Nobutaka Koibuchi
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; (T.F.); (F.S.); (Y.T.); (K.S.); (N.K.); (N.K.)
| | - Kaori Akazawa
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; (K.A.); (Y.K.); (K.S.)
| | - Yuko Kanemoto
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; (K.A.); (Y.K.); (K.S.)
| | - Hidehito Kuroyanagi
- Department of Biochemistry, Graduate School of Medicine, University of the Ryukyu, Okinawa 903-0213, Japan;
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; (K.A.); (Y.K.); (K.S.)
| | - Hiromi Rakugi
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan;
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; (T.F.); (F.S.); (Y.T.); (K.S.); (N.K.); (N.K.)
- Correspondence: ; Tel.: +81-6-6210-8352; Fax: +81-6-6210-8354
| |
Collapse
|
75
|
Sanches SM, Braun AC, Calsavara VF, Barbosa PNVP, Chinen LTD. Comparison of hormonal receptor expression and HER2 status between circulating tumor cells and breast cancer metastases. Clinics (Sao Paulo) 2021; 76:e2971. [PMID: 34644733 PMCID: PMC8478133 DOI: 10.6061/clinics/2021/e2971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/19/2021] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES Breast cancer (BC) is the most common neoplasm in women. Biopsy of metastatic lesions is recommended to confirm estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) status as there are discrepancies in these patterns between primary tumors and metastases in up to 40% of the cases. Circulating tumor cells (CTCs) are related to BC outcomes and could potentially be an alternative to the invasive procedures of metastasis rebiopsy. ISET® technology is not currently employed to detect CTCs in patients with BC. Emerging data support that the characterization of CTC protein expression can refine its prognostic value. Transforming growth factor (TGF)-β plays a role in BC progression and invasiveness. Thus, in this study, we aimed to compare ER, PR, and HER2 expression in primary tumors, CTCs, and metastases and evaluate TGF-β type 1 receptor (TGF-β RI) expression in CTCs as prognostic factor for progression free survival (PFS) and overall survival (OS). METHODS This prospective study was conducted at the A.C. Camargo Cancer Center, Brazil. Blood samples were processed in ISET® (Isolation by SizE of Tumors, Rarecells, France) before computed tomography-guided biopsy of suspected metastatic lesions. Protein expression levels in CTCs were compared to those in primary tumors/metastases (medical records). RESULTS Of the 39 patients initially included, 27 underwent both biopsies of metastases and blood collection and were considered for analysis. The concordance rates for ER, PR, and HER2 expression between primary tumors and metastases were high. No loss of HER2 expression at any metastasis site and retention of the same pattern of protein expression in all triple-negative (TN) tumors (92.5%, 81.5% and 96.2% respectively) (p<0.0001) was observed. When metastases/CTCs were classified as TN/non-TN, CTCs showed high specificity (93%), accuracy (84.2%), and negative predictive value (88%). The median OS of patients without TGF-β RI expression in CTCs was 42.6 versus 20.8 months for TGF-β RI expression-positive ones (p>0.05). CONCLUSION The role of CTCs detected by ISET has not yet been established in BC. Here, we suggest that this methodology may be useful to evaluate metastasis in non-TN cases as well as TGF-β RI expression in CTCs, which may impact patient survival. Due to sample limitations, future studies must focus on specific BC subtypes and an expansion of the cohort.
Collapse
|
76
|
Abstract
ABSTRACT Triple-negative breast cancer (TNBC) is pathologically defined by lack of expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 amplification and portends an aggressive clinical course with worse outcomes compared with other breast cancers. Until recently, standard treatment options consisted of sequential cytotoxic chemotherapies for both early and metastatic disease. Advances in sequencing technology have led to the identification of 4 main subtypes of TNBC based on recurrent genetic alterations, transcriptional patterns, and molecular features: basal-like 1 (BL1), basal-like 2 (BL2), mesenchymal (M), and luminal androgen receptor (LAR). Frequent alterations found in DNA damage response pathways, germline and somatic BRCA1/2 genes, PI3K signaling pathways, and the presence of androgen receptors and infiltrating immune cells could serve as actionable targets to optimize treatments and improve outcomes for patients with TNBC. Recent approvals for immune checkpoint inhibitors and the antibody-drug conjugate, sacituzumab govitecan-hziy, for advanced TNBC illustrate the advances in treatment that can result from these molecular discoveries. This review will explore the molecular subtypes of TNBC and their distinct characteristics, as well as highlight the molecular features and potential "drivers" that have been identified as promising targets for new treatment strategies.
Collapse
|
77
|
Chen Y, Klingen TA, Aas H, Wik E, Akslen LA. Tumor-associated lymphocytes and macrophages are related to stromal elastosis and vascular invasion in breast cancer. J Pathol Clin Res 2021; 7:517-527. [PMID: 34076969 PMCID: PMC8363927 DOI: 10.1002/cjp2.226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/25/2021] [Accepted: 05/05/2021] [Indexed: 02/04/2023]
Abstract
The tumor microenvironment plays a critical role in breast cancer progression. Here, we investigated tumor-infiltrating lymphocytes (TILs) and associations with macrophage numbers, tumor stromal elastosis, vascular invasion, and tumor detection mode. We performed a population-based retrospective study using data from The Norwegian Breast Cancer Screening Program in Vestfold County (2004-2009), including 200 screen-detected and 82 interval cancers. The number of TILs (CD45+, CD3+, CD4+, CD8+, and FOXP3+) and tumor-associated macrophages (CD163+) was counted using immunohistochemistry on tissue microarray slides. Lymphatic and blood vessel invasion (LVI and BVI) were recorded using D2-40 and CD31 staining, and the amount of elastosis (high/low) was determined on regular HE-stained slides. High numbers of all TIL subsets were associated with LVI (p ≤ 0.04 for all), and high counts of several TIL subgroups (CD8+, CD45+, and FOXP3+) were associated with BVI (p ≤ 0.04 for all). Increased levels of all TIL subsets, except CD4+, were associated with estrogen receptor-negative tumors (p < 0.001) and high tumor cell proliferation by Ki67 (p < 0.001). Furthermore, high levels of all TIL subsets were associated with high macrophage counts (p < 0.001) and low-grade stromal elastosis (p ≤ 0.02). High counts of CD3+, CD8+, and FOXP3+ TILs were associated with interval detected tumors (p ≤ 0.04 for all). Finally, in the luminal A subgroup, high levels of CD3+ and FOXP3+ TILs were associated with shorter recurrence-free survival, and high counts of FOXP3+ were linked to reduced breast cancer-specific survival. In conclusion, higher levels of different TIL subsets were associated with stromal features such as high macrophage counts (CD163+), presence of vascular invasion, absence of stromal elastosis, as well as increased tumor cell proliferation and interval detection mode. Our findings support a link between immune cells and vascular invasion in more aggressive breast cancer. Notably, presence of TIL subsets showed prognostic value within the luminal A category.
Collapse
Affiliation(s)
- Ying Chen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of PathologyVestfold HospitalTønsbergNorway
- Department of PathologyOslo University HospitalOsloNorway
| | - Tor Audun Klingen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of PathologyVestfold HospitalTønsbergNorway
| | - Hans Aas
- Department of SurgeryVestfold HospitalTønsbergNorway
| | - Elisabeth Wik
- Centre for Cancer Biomarkers CCBIO, Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of PathologyHaukeland University HospitalBergenNorway
| | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of PathologyHaukeland University HospitalBergenNorway
| |
Collapse
|
78
|
Gawin M, Kurczyk A, Niemiec J, Stanek-Widera A, Grela-Wojewoda A, Adamczyk A, Biskup-Frużyńska M, Polańska J, Widłak P. Intra-Tumor Heterogeneity Revealed by Mass Spectrometry Imaging Is Associated with the Prognosis of Breast Cancer. Cancers (Basel) 2021; 13:4349. [PMID: 34503159 PMCID: PMC8431441 DOI: 10.3390/cancers13174349] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Intra-tumor heterogeneity (ITH) results from the coexistence of genetically distinct cancer cell (sub)populations, their phenotypic plasticity, and the presence of heterotypic components of the tumor microenvironment (TME). Here we addressed the potential association between phenotypic ITH revealed by mass spectrometry imaging (MSI) and the prognosis of breast cancer. Tissue specimens resected from 59 patients treated radically due to the locally advanced HER2-positive invasive ductal carcinoma were included in the study. After the on-tissue trypsin digestion of cellular proteins, peptide maps of all cancer regions (about 380,000 spectra in total) were segmented by an unsupervised approach to reveal their intrinsic heterogeneity. A high degree of similarity between spectra was observed, which indicated the relative homogeneity of cancer regions. However, when the number and diversity of the detected clusters of spectra were analyzed, differences between patient groups were observed. It is noteworthy that a higher degree of heterogeneity was found in tumors from patients who remained disease-free during a 5-year follow-up (n = 38) compared to tumors from patients with progressive disease (distant metastases detected during the follow-up, n = 21). Interestingly, such differences were not observed between patients with a different status of regional lymph nodes, cancer grade, or expression of estrogen receptor at the time of the primary treatment. Subsequently, spectral components with different abundance in cancer regions were detected in patients with different outcomes, and their hypothetical identity was established by assignment to measured masses of tryptic peptides identified in corresponding tissue lysates. Such differentiating components were associated with proteins involved in immune regulation and hemostasis. Further, a positive correlation between the level of tumor-infiltrating lymphocytes and heterogeneity revealed by MSI was observed. We postulate that a higher heterogeneity of tumors with a better prognosis could reflect the presence of heterotypic components including infiltrating immune cells, that facilitated the response to treatment.
Collapse
Affiliation(s)
- Marta Gawin
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.G.); (A.K.); (A.S.-W.); (M.B.-F.)
| | - Agata Kurczyk
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.G.); (A.K.); (A.S.-W.); (M.B.-F.)
| | - Joanna Niemiec
- Maria Skłodowska-Curie National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland; (J.N.); (A.G.-W.); (A.A.)
- Medical College of Rzeszow University, 35-959 Rzeszów, Poland
| | - Agata Stanek-Widera
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.G.); (A.K.); (A.S.-W.); (M.B.-F.)
- Faculty of Medicine, University of Technology in Katowice, 40-555 Katowice, Poland
| | - Aleksandra Grela-Wojewoda
- Maria Skłodowska-Curie National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland; (J.N.); (A.G.-W.); (A.A.)
| | - Agnieszka Adamczyk
- Maria Skłodowska-Curie National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland; (J.N.); (A.G.-W.); (A.A.)
| | - Magdalena Biskup-Frużyńska
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.G.); (A.K.); (A.S.-W.); (M.B.-F.)
| | | | - Piotr Widłak
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.G.); (A.K.); (A.S.-W.); (M.B.-F.)
| |
Collapse
|
79
|
Martínez-López A, García-Casas A, Bragado P, Orimo A, Castañeda-Saucedo E, Castillo-Lluva S. Inhibition of RAC1 activity in cancer associated fibroblasts favours breast tumour development through IL-1β upregulation. Cancer Lett 2021; 521:14-28. [PMID: 34419498 DOI: 10.1016/j.canlet.2021.08.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/11/2021] [Accepted: 08/14/2021] [Indexed: 11/26/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are highly abundant stromal components in the tumour microenvironment. These cells contribute to tumorigenesis and indeed, they have been proposed as a target for anti-cancer therapies. Similarly, targeting the Rho-GTPase RAC1 has also been suggested as a potential therapeutic target in cancer. Here, we show that targeting RAC1 activity, either pharmacologically or by genetic silencing, increases the pro-tumorigenic activity of CAFs by upregulating IL-1β secretion. Moreover, inhibiting RAC1 activity shifts the CAF subtype to a more aggressive phenotype. Thus, as RAC1 suppresses the secretion of IL-1β by CAFs, reducing RAC1 activity in combination with the depletion of this cytokine should be considered as an interesting therapeutic option for breast cancer in which tumour cells retain intact IL-1β signalling.
Collapse
Affiliation(s)
- Angélica Martínez-López
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Biológicas, Universidad Complutense, Madrid, 28040, Spain; Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Mexico
| | - Ana García-Casas
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Biológicas, Universidad Complutense, Madrid, 28040, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, 28040, Spain
| | - Paloma Bragado
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, 28040, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense, Madrid, 28040, Spain
| | - Akira Orimo
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Eduardo Castañeda-Saucedo
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Mexico
| | - Sonia Castillo-Lluva
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Biológicas, Universidad Complutense, Madrid, 28040, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, 28040, Spain.
| |
Collapse
|
80
|
Nordin FJ, Pearanpan L, Chan KM, Kumolosasi E, Yong YK, Shaari K, Rajab NF. Immunomodulatory potential of Clinacanthus nutans extracts in the co-culture of triple-negative breast cancer cells, MDA-MB-231, and THP-1 macrophages. PLoS One 2021; 16:e0256012. [PMID: 34379689 PMCID: PMC8357171 DOI: 10.1371/journal.pone.0256012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer is the main type of breast carcinoma that causes mortality among women because of the limited treatment options and high recurrence. Chronic inflammation has been linked with the tumor microenvironment (TME) in breast cancer progression. Clinacanthus nutans (CN) has gained much attention because of its anticancer properties, but its mechanism remains unclear. We aimed to study the qualitative phytochemical content and elucidate the cytotoxicity effects of CN on human triple-negative breast cancer (TNBC), MDA-MB-231 and human macrophage-like cells such as THP-1 by using sulforhodamine B (SRB) assay. As highly metastatic cells, MDA-MB-231 cells can migrate to the distal position, the effect of CN on migration were also elucidated using the scratch assay. The CN effects on ameliorating chronic inflammation in TME were studied following the co-culture of MDA-MB-231/THP-1 macrophages. The cytokine expression levels of IL-6, IL-1β and tumor necrosis factor-alpha (TNF-α) were determined using ELISA assays. The results showed that both ethanolic and aqueous CN extracts contained alkaloid, phenol and tannin, flavonoid, terpenoid, glycoside and steroid. However, saponin was only found in the aqueous extract of CN. CN was not cytotoxic to both MDA-MB-231 and THP-1 cells. The ability of MDA-MB-231 to migrate was also not halted by CN treatment. However, CN ethanol extract decreased IL-6 at 25 μg/mL (p = 0.02) and 100 μg/mL (p = 0.03) but CN aqueous extract increased IL-6 expression at 50 μg/mL (p = 0.08) and 100 μg/mL (p = 0.02). IL-1β showed decreased expression after treated with CN ethanol and CN aqueous both at 25 μg/mL (p = 0.03). TNF-α were significantly decreased after CN ethanol treatment at concentration 25- (p = 0.001), 50- (p = 0.000) and 100 μg/mL (p = 0.000). CN aqueous extract slightly inhibited TNF-α at all 25–50- and 100 μg/mL (p = 0.001, p = 0.000, p = 0.000, respectively). Overall, CN acts by ameliorating the pro-inflammatory condition in the TME and may be a potential strategy for its anticancer mechanism on highly metastatic breast cancer condition. The major pathways that link both cancer and inflammation were NF-κB and STATs thus further study on the upstream and downstream pathways is needed to fully understand the mechanism of CN extracts in cooling the inflamed TME in breast cancer.
Collapse
Affiliation(s)
- Fariza Juliana Nordin
- Biomedical Science Program, Center for Healthy Aging and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Lishantini Pearanpan
- Biomedical Science Program, Center for Healthy Aging and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kok Meng Chan
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Endang Kumolosasi
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Seri Kembangan, Selangor, Malaysia
| | - Khozirah Shaari
- Faculty of Science, Universiti Putra Malaysia, UPM Serdang, Seri Kembangan, Malaysia
| | - Nor Fadilah Rajab
- Biomedical Science Program, Center for Healthy Aging and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- * E-mail:
| |
Collapse
|
81
|
Del Piccolo N, Shirure VS, Bi Y, Goedegebuure SP, Gholami S, Hughes CC, Fields RC, George SC. Tumor-on-chip modeling of organ-specific cancer and metastasis. Adv Drug Deliv Rev 2021; 175:113798. [PMID: 34015419 DOI: 10.1016/j.addr.2021.05.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 02/08/2023]
Abstract
Every year, cancer claims millions of lives around the globe. Unfortunately, model systems that accurately mimic human oncology - a requirement for the development of more effective therapies for these patients - remain elusive. Tumor development is an organ-specific process that involves modification of existing tissue features, recruitment of other cell types, and eventual metastasis to distant organs. Recently, tissue engineered microfluidic devices have emerged as a powerful in vitro tool to model human physiology and pathology with organ-specificity. These organ-on-chip platforms consist of cells cultured in 3D hydrogels and offer precise control over geometry, biological components, and physiochemical properties. Here, we review progress towards organ-specific microfluidic models of the primary and metastatic tumor microenvironments. Despite the field's infancy, these tumor-on-chip models have enabled discoveries about cancer immunobiology and response to therapy. Future work should focus on the development of autologous or multi-organ systems and inclusion of the immune system.
Collapse
|
82
|
Darbre PD. Endocrine disrupting chemicals and breast cancer cells. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 92:485-520. [PMID: 34452695 DOI: 10.1016/bs.apha.2021.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Many hundreds of endocrine disrupting chemicals (EDCs) have been measured as entering human breast tissue from a range of environmental sources, and this review focuses on discussion of mechanisms by which such EDCs may be contributing to the globally rising incidence of breast cancer. Many of the distinguishing features of breast cancer may be accounted for by EDC exposure, including, but not limited to, the fact that many EDCs possess estrogenic activity and exposure to estrogen is a main risk factor for breast cancer. Studies of the actions of EDCs in human breast cancer cells are aided by use of the conceptual framework of the hallmarks of cancer, and, acting by a variety of genomic and nongenomic mechanisms, EDCs have now been shown to enable all the hallmarks of cancer to develop in human breast cancer cells. Many studies report that hallmarks can develop at concentrations which are within the range of those measured in human breast tissues, especially when added as mixtures. The varied levels of different EDCs measured in individual breast tissue samples together with the overlapping and complementary mechanisms of action of the EDCs imply that thematic mechanisms will be driven inevitably by different chemical mixtures. Despite the complexity, EDCs do need to now be acknowledged as a risk factor for breast cancer in order for preventative strategies to include reduction in EDC exposure.
Collapse
Affiliation(s)
- Philippa D Darbre
- School of Biological Sciences, University of Reading, Reading, United Kingdom.
| |
Collapse
|
83
|
Carter EP, Roozitalab R, Gibson SV, Grose RP. Tumour microenvironment 3D-modelling: simplicity to complexity and back again. Trends Cancer 2021; 7:1033-1046. [PMID: 34312120 DOI: 10.1016/j.trecan.2021.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023]
Abstract
Tumours are surrounded by a host of noncancerous cells that fulfil both supportive and suppressive roles within the tumour microenvironment (TME). The drive to understand the biology behind each of these components has led to a rapid expansion in the number and use of 3D in vitro models, as researchers find ways to incorporate multiple cell types into physiomimetic configurations. The use and increasing complexity of these models does however demand many considerations. In this review we discuss approaches adopted to recapitulate complex tumour biology in tractable 3D models. We consider how these cell types can be sourced and combined and examine methods for the deconvolution of complex multicellular models into manageable and informative outputs.
Collapse
Affiliation(s)
- Edward P Carter
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Reza Roozitalab
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Shayin V Gibson
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
84
|
Ferreira LP, Gaspar VM, Mendes L, Duarte IF, Mano JF. Organotypic 3D decellularized matrix tumor spheroids for high-throughput drug screening. Biomaterials 2021; 275:120983. [PMID: 34186236 DOI: 10.1016/j.biomaterials.2021.120983] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
Decellularized extracellular matrix (dECM) is emerging as a valuable tool for generating 3D in vitro tumor models that better recapitulate tumor-stroma interactions. However, the development of dECM-3D heterotypic microtumors exhibiting a controlled morphology is yet to be materialized. Precisely controlling microtumors morphologic features is key to avoid an inaccurate evaluation of therapeutics performance during preclinical screening. To address this, herein we employed ultra-low adhesion surfaces for bioengineering organotypic 3D metastatic breast cancer-fibroblast models enriched with dECM microfibrillar fragments, as a bottom-up strategy to include major matrix components and their associated biomolecular cues during the early stages of 3D microtissue spheroids assembly, simulating pre-existing ECM presence in the in vivo setting. This biomimetic approach enabled the self-assembly of dECM-3D tumor-stroma spheroids with tunable size and reproducible morphology. Along time, dECM enriched and stroma-rich microtumors exhibited necrotic core formation, secretion of key biomarkers and higher cancer-cell specific resistance to different chemotherapeutics in comparison to standard spheroids. Exometabolomics profiling of dECM-Spheroid in vitro models further identified important breast cancer metabolic features including glucose/pyruvate consumption and lactate excretion, which suggest an intense glycolytic activity, recapitulating major hallmarks of the native microenvironment. Such organotypic dECM-enriched microtumors overcome the morphologic variability generally associated with cell-laden dECM models, while providing a scalable testing platform that can be foreseeable leveraged for high-throughput screening of candidate therapeutics.
Collapse
Affiliation(s)
- Luís P Ferreira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Luís Mendes
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Iola F Duarte
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
85
|
Sharma VP, Williams J, Leung E, Sanders J, Eddy R, Castracane J, Oktay MH, Entenberg D, Condeelis JS. SUN-MKL1 Crosstalk Regulates Nuclear Deformation and Fast Motility of Breast Carcinoma Cells in Fibrillar ECM Microenvironment. Cells 2021; 10:1549. [PMID: 34205257 PMCID: PMC8234170 DOI: 10.3390/cells10061549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022] Open
Abstract
Aligned collagen fibers provide topography for the rapid migration of single tumor cells (streaming migration) to invade the surrounding stroma, move within tumor nests towards blood vessels to intravasate and form distant metastases. Mechanisms of tumor cell motility have been studied extensively in the 2D context, but the mechanistic understanding of rapid single tumor cell motility in the in vivo context is still lacking. Here, we show that streaming tumor cells in vivo use collagen fibers with diameters below 3 µm. Employing 1D migration assays with matching in vivo fiber dimensions, we found a dependence of tumor cell motility on 1D substrate width, with cells moving the fastest and the most persistently on the narrowest 1D fibers (700 nm-2.5 µm). Interestingly, we also observed nuclear deformation in the absence of restricting extracellular matrix pores during high speed carcinoma cell migration in 1D, similar to the nuclear deformation observed in tumor cells in vivo. Further, we found that actomyosin machinery is aligned along the 1D axis and actomyosin contractility synchronously regulates cell motility and nuclear deformation. To further investigate the link between cell speed and nuclear deformation, we focused on the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex proteins and SRF-MKL1 signaling, key regulators of mechanotransduction, actomyosin contractility and actin-based cell motility. Analysis of The Cancer Genome Atlas dataset showed a dramatic decrease in the LINC complex proteins SUN1 and SUN2 in primary tumor compared to the normal tissue. Disruption of LINC complex by SUN1 + 2 KD led to multi-lobular elongated nuclei, increased tumor cell motility and concomitant increase in F-actin, without affecting Lamin proteins. Mechanistically, we found that MKL1, an effector of changes in cellular G-actin to F-actin ratio, is required for increased 1D motility seen in SUN1 + 2 KD cells. Thus, we demonstrate a previously unrecognized crosstalk between SUN proteins and MKL1 transcription factor in modulating nuclear shape and carcinoma cell motility in an in vivo relevant 1D microenvironment.
Collapse
Affiliation(s)
- Ved P. Sharma
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - James Williams
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA; (J.W.); (J.S.); (J.C.)
| | - Edison Leung
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
| | - Joe Sanders
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA; (J.W.); (J.S.); (J.C.)
| | - Robert Eddy
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
| | - James Castracane
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA; (J.W.); (J.S.); (J.C.)
| | - Maja H. Oktay
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David Entenberg
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John S. Condeelis
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
86
|
Zou T, Lu W, Mezhuev Y, Lan M, Li L, Liu F, Cai T, Wu X, Cai Y. A review of nanoparticle drug delivery systems responsive to endogenous breast cancer microenvironment. Eur J Pharm Biopharm 2021; 166:30-43. [PMID: 34098073 DOI: 10.1016/j.ejpb.2021.05.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/24/2021] [Accepted: 05/31/2021] [Indexed: 12/26/2022]
Abstract
Breast cancer, as a malignant disease that seriously threatens women's health, urgently needs to be researched to develop effective and safe therapeutic drugs. Nanoparticle drug delivery systems (NDDS), provide a powerful means for drug targeting to the breast cancer, enhancing the bioavailability and reducing the adverse effects of anticancer drug. However, the breast cancer microenvironment together with heterogeneity of cancer, impedes the tumor targeting effect of NDDS. Breast cancer microenvironment, exerts endogenous stimuli, such as hypoxia, acidosis, and aberrant protease expression, shape a natural shelter for tumor growth, invasion and migration. On the basis of the ubiquitous of endogenous stimuli in the breast cancer microenvironment, researchers exploited them to design the stimuli-responsive NDDS, which response to endogenous stimulus, targeted release drug in breast cancer microenvironment. In this review, we highlighted the effect of the breast cancer microenvironment, summarized innovative NDDS responsive to the internal stimuli in the tumor microenvironment, including the material, the targeting groups, the loading drugs, targeting position and the function of stimuli-responsive nanoparticle drug delivery system. The limitations and potential applications of the stimuli-responsive nanoparticle drug delivery systems for breast cancer treatment were discussed to further the application.
Collapse
Affiliation(s)
- Tengteng Zou
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Wenping Lu
- Guang an'men Hospital China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yaroslav Mezhuev
- Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russia
| | - Meng Lan
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Lihong Li
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Fengjie Liu
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Tiange Cai
- College of Life Sciences, Liaoning University, Shenyang 110036, PR China.
| | - Xiaoyu Wu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada.
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China; Guangdong Key Lab of Traditional Chinese Medicine Information Technology, Jinan University, Guangzhou 510632, PR China; Cancer Research Institute, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
87
|
Na D, Moon HG. Patient-Derived Xenograft Models in Breast Cancer Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1187:283-301. [PMID: 33983584 DOI: 10.1007/978-981-32-9620-6_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Patient-derived xenograft (PDX) model can be used as a platform to study the individual patient's sensitivity to targeted agents as well as its ability to guide our understanding in various aspects of tumor biology including the tumor's clonal evolution and interaction with microenvironment. In this chapter, we review the history of PDX models in various tumor types. Additionally, we highlight the key studies that suggested potential value of PDX models in cancer treatment. Specifically, we will briefly introduce several studies on the issue of PDX models for precision medicine. In latter part of this chapter, we focus on the studies that used PDX models to investigate the molecular biology of breast cancer that underlies the process of drug resistance and tumor metastasis. Also, we will address our own experience in developing PDX models using breast cancer tissues from Korean breast cancer patients.
Collapse
Affiliation(s)
- Deukchae Na
- Institute of Convergence Medicine, Ewha Womans University Mokdong Hospital, Seoul, South Korea
| | - Hyeong-Gon Moon
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
88
|
Abubakar M, Zhang J, Ahearn TU, Koka H, Guo C, Lawrence SM, Mutreja K, Figueroa JD, Ying J, Lissowska J, Lyu N, Garcia-Closas M, Yang XR. Tumor-Associated Stromal Cellular Density as a Predictor of Recurrence and Mortality in Breast Cancer: Results from Ethnically Diverse Study Populations. Cancer Epidemiol Biomarkers Prev 2021; 30:1397-1407. [PMID: 33952648 DOI: 10.1158/1055-9965.epi-21-0055] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/08/2021] [Accepted: 04/26/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Tumor-associated stroma is comprised of fibroblasts, tumor-infiltrating lymphocytes (TIL), macrophages, endothelial cells, and other cells that interactively influence tumor progression through inflammation and wound repair. Although gene-expression signatures reflecting wound repair predict breast cancer survival, it is unclear whether combined density of tumor-associated stromal cells, a morphologic proxy for inflammation and wound repair signatures on routine hematoxylin and eosin (H&E)-stained sections, is of prognostic relevance. METHODS By applying machine learning to digitized H&E-stained sections for 2,084 breast cancer patients from China (n = 596; 24-55 years), Poland (n = 810; 31-75 years), and the United States (n = 678; 55-78 years), we characterized tumor-associated stromal cellular density (SCD) as the percentage of tumor-stroma that is occupied by nucleated cells. Hazard ratios (HR) and 95% confidence intervals (CI) for associations between SCD and clinical outcomes [recurrence (China) and mortality (Poland and the United States)] were estimated using Cox proportional hazard regression, adjusted for clinical variables. RESULTS SCD was independently predictive of poor clinical outcomes in hormone receptor-positive (luminal) tumors from China [multivariable HR (95% CI)fourth(Q4) vs. first(Q1) quartile = 1.86 (1.06-3.26); P trend = 0.03], Poland [HR (95% CI)Q4 vs. Q1 = 1.80 (1.12-2.89); P trend = 0.01], and the United States [HR (95% CI)Q4 vs. Q1 = 2.42 (1.33-4.42); P trend = 0.002]. In general, SCD provided more prognostic information than most classic clinicopathologic factors, including grade, size, PR, HER2, IHC4, and TILs, predicting clinical outcomes irrespective of menopausal or lymph nodal status. SCD was not predictive of outcomes in hormone receptor-negative tumors. CONCLUSIONS Our findings support the independent prognostic value of tumor-associated SCD among ethnically diverse luminal breast cancer patients. IMPACT Assessment of tumor-associated SCD on standard H&E could help refine prognostic assessment and therapeutic decision making in luminal breast cancer.
Collapse
Affiliation(s)
- Mustapha Abubakar
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland.
| | - Jing Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Thomas U Ahearn
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Hela Koka
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Changyuan Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Scott M Lawrence
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Karun Mutreja
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Jonine D Figueroa
- Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jolanta Lissowska
- Epidemiology Unit, Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Ning Lyu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Montserrat Garcia-Closas
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Xiaohong Rose Yang
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| |
Collapse
|
89
|
Adams SD, Csere J, D'angelo G, Carter EP, Romao M, Arnandis T, Dodel M, Kocher HM, Grose R, Raposo G, Mardakheh F, Godinho SA. Centrosome amplification mediates small extracellular vesicle secretion via lysosome disruption. Curr Biol 2021; 31:1403-1416.e7. [PMID: 33592190 PMCID: PMC8047808 DOI: 10.1016/j.cub.2021.01.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/01/2020] [Accepted: 01/08/2021] [Indexed: 01/08/2023]
Abstract
Bidirectional communication between cells and their surrounding environment is critical in both normal and pathological settings. Extracellular vesicles (EVs), which facilitate the horizontal transfer of molecules between cells, are recognized as an important constituent of cell-cell communication. In cancer, alterations in EV secretion contribute to the growth and metastasis of tumor cells. However, the mechanisms underlying these changes remain largely unknown. Here, we show that centrosome amplification is associated with and sufficient to promote small extracellular vesicle (SEV) secretion in pancreatic cancer cells. This is a direct result of lysosomal dysfunction, caused by increased reactive oxygen species (ROS) downstream of extra centrosomes. We propose that defects in lysosome function could promote multivesicular body fusion with the plasma membrane, thereby enhancing SEV secretion. Furthermore, we find that SEVs secreted in response to amplified centrosomes are functionally distinct and activate pancreatic stellate cells (PSCs). These activated PSCs promote the invasion of pancreatic cancer cells in heterotypic 3D cultures. We propose that SEVs secreted by cancer cells with amplified centrosomes influence the bidirectional communication between the tumor cells and the surrounding stroma to promote malignancy.
Collapse
Affiliation(s)
- Sophie D Adams
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Judit Csere
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Gisela D'angelo
- Structure and Membrane Compartments, Institute Curie, Paris Sciences & Lettres Research University, Centre for National de la Recherche Scientifique, UMR144, Paris, France
| | - Edward P Carter
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Maryse Romao
- Structure and Membrane Compartments, Institute Curie, Paris Sciences & Lettres Research University, Centre for National de la Recherche Scientifique, UMR144, Paris, France
| | - Teresa Arnandis
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Martin Dodel
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Hemant M Kocher
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Richard Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Graça Raposo
- Structure and Membrane Compartments, Institute Curie, Paris Sciences & Lettres Research University, Centre for National de la Recherche Scientifique, UMR144, Paris, France
| | - Faraz Mardakheh
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Susana A Godinho
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
90
|
Plasmin and Plasminogen System in the Tumor Microenvironment: Implications for Cancer Diagnosis, Prognosis, and Therapy. Cancers (Basel) 2021; 13:cancers13081838. [PMID: 33921488 PMCID: PMC8070608 DOI: 10.3390/cancers13081838] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In this review, we present a detailed discussion of how the plasminogen-activation system is utilized by tumor cells in their unrelenting attack on the tissues surrounding them. Plasmin is an enzyme which is responsible for digesting several proteins that hold the tissues surrounding solid tumors together. In this process tumor cells utilize the activity of plasmin to digest tissue barriers in order to leave the tumour site and spread to other parts of the body. We specifically focus on the role of plasminogen receptor—p11 which is an important regulatory protein that facilitates the conversion of plasminogen to plasmin and by this means promotes the attack by the tumour cells on their surrounding tissues. Abstract The tumor microenvironment (TME) is now being widely accepted as the key contributor to a range of processes involved in cancer progression from tumor growth to metastasis and chemoresistance. The extracellular matrix (ECM) and the proteases that mediate the remodeling of the ECM form an integral part of the TME. Plasmin is a broad-spectrum, highly potent, serine protease whose activation from its precursor plasminogen is tightly regulated by the activators (uPA, uPAR, and tPA), the inhibitors (PAI-1, PAI-2), and plasminogen receptors. Collectively, this system is called the plasminogen activation system. The expression of the components of the plasminogen activation system by malignant cells and the surrounding stromal cells modulates the TME resulting in sustained cancer progression signals. In this review, we provide a detailed discussion of the roles of plasminogen activation system in tumor growth, invasion, metastasis, and chemoresistance with specific emphasis on their role in the TME. We particularly review the recent highlights of the plasminogen receptor S100A10 (p11), which is a pivotal component of the plasminogen activation system.
Collapse
|
91
|
Fulfager AD, Yadav KS. Understanding the implications of co-delivering therapeutic agents in a nanocarrier to combat multidrug resistance (MDR) in breast cancer. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102405] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
92
|
Malherbe K. Tumor Microenvironment and the Role of Artificial Intelligence in Breast Cancer Detection and Prognosis. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1364-1373. [PMID: 33639101 DOI: 10.1016/j.ajpath.2021.01.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/02/2021] [Accepted: 01/28/2021] [Indexed: 12/21/2022]
Abstract
A critical knowledge gap has been noted in breast cancer detection, prognosis, and evaluation between tumor microenvironment and associated neoplasm. Artificial intelligence (AI) has multiple subsets or methods for data extraction and evaluation, including artificial neural networking, which allows computational foundations, similar to neurons, to make connections and new neural pathways during data set training. Deep machine learning and AI hold great potential to accurately assess tumor microenvironment models employing vast data management techniques. Despite the significant potential AI holds, there is still much debate surrounding the appropriate and ethical curation of medical data from picture archiving and communication systems. AI output's clinical significance depends on its human predecessor's data training sets. Integration between biomarkers, risk factors, and imaging data will allow the best predictor models for patient-based outcomes.
Collapse
Affiliation(s)
- Kathryn Malherbe
- Department Radiography, Faculty Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
93
|
Stromal CCL5 Promotes Breast Cancer Progression by Interacting with CCR3 in Tumor Cells. Int J Mol Sci 2021; 22:ijms22041918. [PMID: 33671956 PMCID: PMC7919043 DOI: 10.3390/ijms22041918] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Chemokines secreted from stromal cells have important roles for interactions with carcinoma cells and regulating tumor progression. C-C motif chemokine ligand (CCL) 5 is expressed in various types of stromal cells and associated with tumor progression, interacting with C-C chemokine receptor (CCR) 1, 3 and 5 expressed in tumor cells. However, the expression on CCL5 and its receptors have so far not been well-examined in human breast carcinoma tissues. We therefore immunolocalized CCL5, as well as CCR1, 3 and 5, in 111 human breast carcinoma tissues and correlated them with clinicopathological characteristics. Stromal CCL5 immunoreactivity was significantly correlated with the aggressive phenotype of breast carcinomas. Importantly, this tendency was observed especially in the CCR3-positive group. Furthermore, the risk of recurrence was significantly higher in the patients with breast carcinomas positive for CCL5 and CCR3 but negative for CCR1 and CCR5, as compared with other patients. In summary, the CCL5-CCR3 axis might contribute to a worse prognosis in breast cancer patients, and these findings will contribute to a better understanding of the significance of the CCL5/CCRs axis in breast carcinoma microenvironment.
Collapse
|
94
|
Miklikova S, Trnkova L, Plava J, Bohac M, Kuniakova M, Cihova M. The Role of BRCA1/2-Mutated Tumor Microenvironment in Breast Cancer. Cancers (Basel) 2021; 13:575. [PMID: 33540843 PMCID: PMC7867315 DOI: 10.3390/cancers13030575] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
Taking into account the factors of high incidence rate, prevalence and mortality, breast cancer represents a crucial social and economic burden. Most cases of breast cancer develop as a consequence of somatic mutations accumulating in mammary epithelial cells throughout lifetime and approximately 5-10% can be ascribed to monogenic predispositions. Even though the role of genetic predispositions in breast cancer is well described in the context of genetics, very little is known about the role of the microenvironment carrying the same aberrant cells impaired by the germline mutation in the breast cancer development and progression. Based on the clinical observations, carcinomas carrying mutations in hereditary tumor-suppressor genes involved in maintaining genome integrity such as BRCA1/2 have worse prognosis and aggressive behavior. One of the mechanisms clarifying the aggressive nature of BRCA-associated tumors implies alterations within the surrounding adipose tissue itself. The objective of this review is to look at the role of BRCA1/2 mutations in the context of breast tumor microenvironment and plausible mechanisms by which it contributes to the aggressive behavior of the tumor cells.
Collapse
Affiliation(s)
- Svetlana Miklikova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| | - Lenka Trnkova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| | - Jana Plava
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| | - Martin Bohac
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, National Cancer Institute, Klenova 1, 83310 Bratislava, Slovakia;
- Department of Oncosurgery, National Cancer Institute, Klenova 1, 83310 Bratislava, Slovakia
- Regenmed Ltd., Medena 29, 81108 Bratislava, Slovakia
| | - Marcela Kuniakova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 81108 Bratislava, Slovakia;
| | - Marina Cihova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| |
Collapse
|
95
|
Dong X, Lv S, Gu D, Zhang X, Ye Z. Up-regulation of L Antigen Family Member 3 Associates With Aggressive Progression of Breast Cancer. Front Oncol 2021; 10:553628. [PMID: 33552947 PMCID: PMC7858652 DOI: 10.3389/fonc.2020.553628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
The role of L Antigen Family Member 3 (LAGE3) in breast cancer (BC) has not been sufficiently studied. In this study, we explored the clinical value and biological functions of LAGE3 in BC. Comprehensive analysis of LAGE3 was carried out on The Cancer Genome Atlas, Molecular Taxonomy of Breast Cancer International Consortium and Gene Expression Omnibus datasets. Results showed that LAGE3 expression was higher in BC tissues than in normal breast tissues of public datasets and our local cohort. Moreover, its expression was higher in BC patients with larger tumor size, significant lymph node metastasis, higher tumor grade, and more advanced disease stage. High expression of LAGE3 was correlated with poor prognosis, and LAGE3 could independently predict survival of BC patients. Functional enrichment analysis revealed a correlation between LAGE3 expression and biochemical metabolism and immune-related terms and cancer-related pathways. Analysis of tumor microenvironment indicated that LAGE3 expression was associated with the immune cell infiltration and anti-cancer immunity cycle. LAGE3 expression was higher in triple-negative breast cancer (TNBC) compared to hormone receptor-positive BC, but not HER2-positive subtype. Suppression of LAGE3 expression inhibited the proliferation and induced apoptosis of TNBC cell lines. Besides, the down-regulation of LAGE3 attenuated the migration and invasion but reduced the expression level of epithelial-mesenchymal-transition related proteins in TNBC cell lines. In conclusion, this study demonstrated for the first time that LAGE3 promotes the progression of BC. Therefore, it may be a potential diagnostic and prognostic biomarker, as well as a treatment target for BC.
Collapse
Affiliation(s)
- Xubin Dong
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shihui Lv
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dianna Gu
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaohua Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhiqiang Ye
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
96
|
Jo SH, Heo WH, Son HY, Quan M, Hong BS, Kim JH, Lee HB, Han W, Park Y, Lee DS, Kwon NH, Park MC, Chae J, Kim JI, Noh DY, Moon HG. S100A8/A9 mediate the reprograming of normal mammary epithelial cells induced by dynamic cell-cell interactions with adjacent breast cancer cells. Sci Rep 2021; 11:1337. [PMID: 33446797 PMCID: PMC7809201 DOI: 10.1038/s41598-020-80625-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
To understand the potential effects of cancer cells on surrounding normal mammary epithelial cells, we performed direct co-culture of non-tumorigenic mammary epithelial MCF10A cells and various breast cancer cells. Firstly, we observed dynamic cell-cell interactions between the MCF10A cells and breast cancer cells including lamellipodia or nanotube-like contacts and transfer of extracellular vesicles. Co-cultured MCF10A cells exhibited features of epithelial-mesenchymal transition, and showed increased capacity of cell proliferation, migration, colony formation, and 3-dimensional sphere formation. Direct co-culture showed most distinct phenotype changes in MCF10A cells followed by conditioned media treatment and indirect co-culture. Transcriptome analysis and phosphor-protein array suggested that several cancer-related pathways are significantly dysregulated in MCF10A cells after the direct co-culture with breast cancer cells. S100A8 and S100A9 showed distinct up-regulation in the co-cultured MCF10A cells and their microenvironmental upregulation was also observed in the orthotropic xenograft of syngeneic mouse mammary tumors. When S100A8/A9 overexpression was induced in MCF10A cells, the cells showed phenotypic features of directly co-cultured MCF10A cells in terms of in vitro cell behaviors and signaling activities suggesting a S100A8/A9-mediated transition program in non-tumorigenic epithelial cells. This study suggests the possibility of dynamic cell-cell interactions between non-tumorigenic mammary epithelial cells and breast cancer cells that could lead to a substantial transition in molecular and functional characteristics of mammary epithelial cells.
Collapse
Affiliation(s)
- Seol Hwa Jo
- Interdisciplinary Graduate Program in Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Woo Hang Heo
- Interdisciplinary Graduate Program in Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Hye-Youn Son
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Mingji Quan
- Interdisciplinary Graduate Program in Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Bok Sil Hong
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Ju Hee Kim
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Han-Byoel Lee
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Surgery, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Wonshik Han
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Surgery, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yeonju Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Nam Hoon Kwon
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, Korea
| | - Min Chul Park
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, Korea
| | - Jeesoo Chae
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Biochemistry, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Il Kim
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Biochemistry, Seoul National University College of Medicine, Seoul, Korea
| | - Dong-Young Noh
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Surgery, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyeong-Gon Moon
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea. .,Department of Surgery, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea. .,Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
97
|
Delort L, Cholet J, Decombat C, Vermerie M, Dumontet C, Castelli FA, Fenaille F, Auxenfans C, Rossary A, Caldefie-Chezet F. The Adipose Microenvironment Dysregulates the Mammary Myoepithelial Cells and Could Participate to the Progression of Breast Cancer. Front Cell Dev Biol 2021; 8:571948. [PMID: 33505957 PMCID: PMC7829501 DOI: 10.3389/fcell.2020.571948] [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: 06/12/2020] [Accepted: 11/25/2020] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most common cancer among women worldwide. Overweight and obesity are now recognized as established risk factors for this pathology in postmenopausal women. These conditions are also believed to be responsible for higher recurrence and mortality rates. Reciprocal interactions have been described between adipose and cancer cells. An adipose microenvironment favors a greater proliferation of cancer cells, their invasion and even resistance to anti-cancer treatments. In addition, the chronic low-grade inflammation observed in obese individuals is believed to amplify these processes. Among the cell types present in the breast, myoepithelial cells (MECs), located at the interface of the epithelial cells and the stroma, are considered "tumor suppressor" cells. During the transition from ductal carcinoma in situ to invasive cancer, disorganization or even the disappearance of MECs is observed, thereby enhancing the ability of the cancer cells to migrate. As the adipose microenvironment is now considered as a central actor in the progression of breast cancer, our objective was to evaluate if it could be involved in MEC functional modifications, leading to the transition of in situ to invasive carcinoma, particularly in obese patients. Through a co-culture model, we investigated the impact of human adipose stem cells from women of normal weight and obese women, differentiated or not into mature adipocytes, on the functionality of the MECs by measuring changes in viability, apoptosis, gene, and miRNA expressions. We found that adipose cells (precursors and differentiated adipocytes) could decrease the viability of the MECs, regardless of the original BMI. The adipose cells could also disrupt the expression of the genes involved in the maintenance of the extracellular matrix and to amplify the expression of leptin and inflammatory markers. miR-122-5p and miR-132-3p could also be considered as targets for adipose cells. The metabolite analyses revealed specific profiles that may be involved in the growth of neoplastic cells. All of these perturbations could thus be responsible for the loss of tumor suppressor status of MECs and promote the transition from in situ to invasive carcinoma.
Collapse
Affiliation(s)
- Laetitia Delort
- Université Clermont Auvergne, INRAE, UNH, ECREIN, Clermont-Ferrand, France
| | - Juliette Cholet
- Université Clermont Auvergne, INRAE, UNH, ECREIN, Clermont-Ferrand, France
| | - Caroline Decombat
- Université Clermont Auvergne, INRAE, UNH, ECREIN, Clermont-Ferrand, France
| | - Marion Vermerie
- Université Clermont Auvergne, INRAE, UNH, ECREIN, Clermont-Ferrand, France
| | - Charles Dumontet
- Université Lyon 1, INSERM U1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
| | - Florence A Castelli
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Gif-sur-Yvette, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Gif-sur-Yvette, France
| | - Céline Auxenfans
- Banque de Tissus et de Cellules, Hôpital Edouard-Herriot, Lyon, France
| | - Adrien Rossary
- Université Clermont Auvergne, INRAE, UNH, ECREIN, Clermont-Ferrand, France
| | | |
Collapse
|
98
|
Amirghasemi F, Adjei-Sowah E, Pockaj BA, Nikkhah M. Microengineered 3D Tumor Models for Anti-Cancer Drug Discovery in Female-Related Cancers. Ann Biomed Eng 2021; 49:1943-1972. [PMID: 33403451 DOI: 10.1007/s10439-020-02704-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
The burden of cancer continues to increase in society and negatively impacts the lives of numerous patients. Due to the high cost of current treatment strategies, there is a crucial unmet need to develop inexpensive preclinical platforms to accelerate the process of anti-cancer drug discovery to improve outcomes in cancer patients, most especially in female patients. Many current methods employ expensive animal models which not only present ethical concerns but also do not often accurately predict human physiology and the outcomes of anti-cancer drug responsiveness. Conventional treatment approaches for cancer generally include systemic therapy after a surgical procedure. Although this treatment technique is effective, the outcome is not always positive due to various complex factors such as intratumor heterogeneity and confounding factors within the tumor microenvironment (TME). Patients who develop metastatic disease still have poor prognosis. To that end, recent efforts have attempted to use 3D microengineered platforms to enhance the predictive power and efficacy of anti-cancer drug screening, ultimately to develop personalized therapies. Fascinating features of microengineered assays, such as microfluidics, have led to the advancement in the development of the tumor-on-chip technology platforms, which have shown tremendous potential for meaningful and physiologically relevant anti-cancer drug discovery and screening. Three dimensional microscale models provide unprecedented ability to unveil the biological complexities of cancer and shed light into the mechanism of anti-cancer drug resistance in a timely and resource efficient manner. In this review, we discuss recent advances in the development of microengineered tumor models for anti-cancer drug discovery and screening in female-related cancers. We specifically focus on female-related cancers to draw attention to the various approaches being taken to improve the survival rate of women diagnosed with cancers caused by sex disparities. We also briefly discuss other cancer types like colon adenocarcinomas and glioblastoma due to their high rate of occurrence in females, as well as the high likelihood of sex-biased mutations which complicate current treatment strategies for women. We highlight recent advances in the development of 3D microscale platforms including 3D tumor spheroids, microfluidic platforms as well as bioprinted models, and discuss how they have been utilized to address major challenges in the process of drug discovery, such as chemoresistance, intratumor heterogeneity, drug toxicity, etc. We also present the potential of these platform technologies for use in high-throughput drug screening approaches as a replacements of conventional assays. Within each section, we will provide our perspectives on advantages of the discussed platform technologies.
Collapse
Affiliation(s)
- Farbod Amirghasemi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287-9709, USA
| | - Emmanuela Adjei-Sowah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287-9709, USA
| | - Barbara A Pockaj
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic, Phoenix, AZ, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287-9709, USA. .,Biodesign Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
99
|
Terkelsen T, Pernemalm M, Gromov P, Børresen-Dale AL, Krogh A, Haakensen VD, Lethiö J, Papaleo E, Gromova I. High-throughput proteomics of breast cancer interstitial fluid: identification of tumor subtype-specific serologically relevant biomarkers. Mol Oncol 2021; 15:429-461. [PMID: 33176066 PMCID: PMC7858121 DOI: 10.1002/1878-0261.12850] [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: 01/22/2020] [Revised: 08/13/2020] [Accepted: 11/09/2020] [Indexed: 12/24/2022] Open
Abstract
Despite significant advancements in breast cancer (BC) research, clinicians lack robust serological protein markers for accurate diagnostics and tumor stratification. Tumor interstitial fluid (TIF) accumulates aberrantly externalized proteins within the local tumor space, which can potentially gain access to the circulatory system. As such, TIF may represent a valuable starting point for identifying relevant tumor-specific serological biomarkers. The aim of the study was to perform comprehensive proteomic profiling of TIF to identify proteins associated with BC tumor status and subtype. A liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis of 35 TIFs of three main subtypes: luminal (19), Her2 (4), and triple-negative (TNBC) (12) resulted in the identification of > 8800 proteins. Unsupervised hierarchical clustering segregated the TIF proteome into two major clusters, luminal and TNBC/Her2 subgroups. High-grade tumors enriched with tumor infiltrating lymphocytes (TILs) were also stratified from low-grade tumors. A consensus analysis approach, including differential abundance analysis, selection operator regression, and random forest returned a minimal set of 24 proteins associated with BC subtypes, receptor status, and TIL scoring. Among them, a panel of 10 proteins, AGR3, BCAM, CELSR1, MIEN1, NAT1, PIP4K2B, SEC23B, THTPA, TMEM51, and ULBP2, was found to stratify the tumor subtype-specific TIFs. In particular, upregulation of BCAM and CELSR1 differentiates luminal subtypes, while upregulation of MIEN1 differentiates Her2 subtypes. Immunohistochemistry analysis showed a direct correlation between protein abundance in TIFs and intratumor expression levels for all 10 proteins. Sensitivity and specificity were estimated for this protein panel by using an independent, comprehensive breast tumor proteome dataset. The results of this analysis strongly support our data, with eight of the proteins potentially representing biomarkers for stratification of BC subtypes. Five of the most representative proteomics databases currently available were also used to estimate the potential for these selected proteins to serve as putative serological markers.
Collapse
Affiliation(s)
- Thilde Terkelsen
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Maria Pernemalm
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Pavel Gromov
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Anna-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway
| | - Anders Krogh
- Department of Computer Science, University of Copenhagen, Denmark.,Department of Biology, University of Copenhagen, Denmark
| | - Vilde D Haakensen
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway
| | - Janne Lethiö
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark.,Translational Disease System Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Denmark
| | - Irina Gromova
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| |
Collapse
|
100
|
Caponnetto F, Dalla E, Mangoni D, Piazza S, Radovic S, Ius T, Skrap M, Di Loreto C, Beltrami AP, Manini I, Cesselli D. The miRNA Content of Exosomes Released from the Glioma Microenvironment Can Affect Malignant Progression. Biomedicines 2020; 8:biomedicines8120564. [PMID: 33287106 PMCID: PMC7761654 DOI: 10.3390/biomedicines8120564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Low-grade gliomas (LGG) are infiltrative primary brain tumors that in 70% of the cases undergo anaplastic transformation, deeply affecting prognosis. However, the timing of progression is heterogeneous. Recently, the tumor microenvironment (TME) has gained much attention either as prognostic factor or therapeutic target. Through the release of extracellular vesicles, the TME contributes to tumor progression by transferring bioactive molecules such as microRNA. The aim of the study was to take advantage of glioma-associated stem cells (GASC), an in vitro model of the glioma microenvironment endowed with a prognostic significance, and their released exosomes, to investigate the possible role of exosome miRNAs in favoring the anaplastic transformation of LGG. Therefore, by deep sequencing, we analyzed and compared the miRNA profile of GASC and exosomes obtained from LGG patients characterized by different prognosis. Results showed that exosomes presented a different signature, when compared to their cellular counterpart and that, although sharing several miRNAs, exosomes of patients with a bad prognosis, selectively expressed some miRNAs possibly responsible for the more aggressive phenotype. These findings get insights into the value of TME and exosomes as potential biomarkers for precision medicine approaches aimed at improving LGG prognostic stratification and therapeutic strategies.
Collapse
Affiliation(s)
- Federica Caponnetto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (E.D.); (C.D.L.); (A.P.B.); (D.C.)
- Correspondence: (F.C.); (I.M.); Tel.: +39-0432-559-412 (F.C. & I.M.)
| | - Emiliano Dalla
- Department of Medicine, University of Udine, 33100 Udine, Italy; (E.D.); (C.D.L.); (A.P.B.); (D.C.)
| | - Damiano Mangoni
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy;
| | - Silvano Piazza
- International Center for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy;
| | | | - Tamara Ius
- Neurosurgery Unit, Department of Neurosciences, University Hospital of Udine, 33100 Udine, Italy; (T.I.); (M.S.)
| | - Miran Skrap
- Neurosurgery Unit, Department of Neurosciences, University Hospital of Udine, 33100 Udine, Italy; (T.I.); (M.S.)
| | - Carla Di Loreto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (E.D.); (C.D.L.); (A.P.B.); (D.C.)
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy
| | - Antonio Paolo Beltrami
- Department of Medicine, University of Udine, 33100 Udine, Italy; (E.D.); (C.D.L.); (A.P.B.); (D.C.)
| | - Ivana Manini
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy
- Correspondence: (F.C.); (I.M.); Tel.: +39-0432-559-412 (F.C. & I.M.)
| | - Daniela Cesselli
- Department of Medicine, University of Udine, 33100 Udine, Italy; (E.D.); (C.D.L.); (A.P.B.); (D.C.)
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy
| |
Collapse
|