1
|
Guo R, Wang P. The complex role of regulatory cells in breast cancer: implication for immunopathogenesis and immunotherapy. Breast Cancer 2025; 32:227-241. [PMID: 39589625 DOI: 10.1007/s12282-024-01654-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
Breast cancers (BCs) are frequently linked to an immunosuppressive microenvironment that facilitates tumor evasion of anti-cancer immunity. The cells that suppress the immune system such as regulatory B cells (Bregs), regulatory T cells (Tregs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), play a crucial role in immune resistance. Also, tumor progression and immune evasion of cancers are facilitated by cytokines and factors released by tumor cells or immunosuppressive cells. Targeting these regulatory cells therapeutically, whether through elimination, inactivation, or reprogramming, has resulted in hopeful anti-tumor reactions. Yet, the substantial diversity and adaptability of these cells, both in terms of appearance and function, as well as their variation over time and depending on where they are in the body, have posed significant challenges for using them as reliable biomarkers and creating focused therapies that could target their creation, growth, and various tumor-promoting roles. The immunotherapy approaches in BC and their effectiveness in treating certain subtypes are still in their initial phases. In this review, we thoroughly outlined the characteristics, roles, and possible treatment options for these immune-suppressing cells in the tumor environment.
Collapse
Affiliation(s)
- RuiJuan Guo
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Shandong Province, Yantai City, People's Republic of China
| | - Ping Wang
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Shandong Province, Yantai City, People's Republic of China.
| |
Collapse
|
2
|
Seitz J, Bilsland A, Puget C, Baasner I, Klopfleisch R, Stein T. SFRP1 Expression is Inversely Associated With Metastasis Formation in Canine Mammary Tumours. J Mammary Gland Biol Neoplasia 2023; 28:15. [PMID: 37402051 DOI: 10.1007/s10911-023-09543-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/22/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Canine mammary tumours (CMTs) are the most frequent tumours in intact female dogs and show strong similarities with human breast cancer. In contrast to the human disease there are no standardised diagnostic or prognostic biomarkers available to guide treatment. We recently identified a prognostic 18-gene RNA signature that could stratify human breast cancer patients into groups with significantly different risk of distant metastasis formation. Here, we assessed whether expression patterns of these RNAs were also associated with canine tumour progression. METHOD A sequential forward feature selection process was performed on a previously published microarray dataset of 27 CMTs with and without lymph node (LN) metastases to identify RNAs with significantly differential expression to identify prognostic genes within the 18-gene signature. Using an independent set of 33 newly identified archival CMTs, we compared expression of the identified prognostic subset on RNA and protein basis using RT-qPCR and immunohistochemistry on FFPE-tissue sections. RESULTS While the 18-gene signature as a whole did not have any prognostic power, a subset of three RNAs: Col13a1, Spock2, and Sfrp1, together completely separated CMTs with and without LN metastasis in the microarray set. However, in the new independent set assessed by RT-qPCR, only the Wnt-antagonist Sfrp1 showed significantly increased mRNA abundance in CMTs without LN metastases on its own (p = 0.013) in logistic regression analysis. This correlated with stronger SFRP1 protein staining intensity of the myoepithelium and/or stroma (p < 0.001). SFRP1 staining, as well as β-catenin membrane staining, was significantly associated with negative LN status (p = 0.010 and 0.014 respectively). However, SFRP1 did not correlate with β-catenin membrane staining (p = 0.14). CONCLUSION The study identified SFRP1 as a potential biomarker for metastasis formation in CMTs, but lack of SFRP1 was not associated with reduced membrane-localisation of β-catenin in CMTs.
Collapse
Affiliation(s)
- Judith Seitz
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Alan Bilsland
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of MVLS, University of Glasgow, Glasgow, UK
| | - Chloé Puget
- Institute of Veterinary Pathology, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Ian Baasner
- Institute of Veterinary Pathology, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Torsten Stein
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Transcriptomic Profile of Canine Mammary Ductal Carcinoma. Int J Mol Sci 2023; 24:ijms24065212. [PMID: 36982287 PMCID: PMC10049542 DOI: 10.3390/ijms24065212] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 03/11/2023] Open
Abstract
Dogs can be excellent models for spontaneous studies about breast cancers, presenting similarities in clinical behavior and molecular pathways of the disease. Thus, analyses of the canine transcriptome can identify deregulated genes and pathways, contributing to the identification of biomarkers and new therapeutic targets, benefiting humans and animals. In this context, this study aimed to determine the transcriptional profile of canine mammary ductal carcinoma and contribute to the clarification of the importance of deregulated molecules in the molecular pathways involved in the disease. Therefore, we used mammary ductal carcinoma tissue samples and non-tumor mammary tissue from the radical mastectomy of six female dogs. Sequencing was performed on the NextSeq-500 System platform. A comparison of carcinoma tissue and normal tissue revealed 633 downregulated and 573 upregulated genes, which were able to differentiate the groups by principal component analysis. Gene ontology analysis indicated that inflammatory, cell differentiation and adhesion, and extracellular matrix maintenance pathways were mainly deregulated in this series. The main differentially expressed genes observed in this research can indicate greater disease aggressiveness and worse prognosis. Finally, the study of the canine transcriptome indicates that it is an excellent model to generate information relevant to oncology in both species.
Collapse
|
4
|
Zhang Z, Bassale S, Jindal S, Fraser A, Guinto E, Anderson W, Mori M, Smith KR, Schedin P. Young-Onset Breast Cancer Outcomes by Time Since Recent Childbirth in Utah. JAMA Netw Open 2022; 5:e2236763. [PMID: 36239933 PMCID: PMC9568799 DOI: 10.1001/jamanetworkopen.2022.36763] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Breast cancer diagnosed within 5 to 10 years after childbirth, called postpartum breast cancer (PPBC), is associated with increased risk for metastasis and death. Whether a postpartum diagnosis is an independent risk factor or a surrogate marker of cancer features associated with poor outcomes remains understudied. OBJECTIVE To determine whether diagnostic temporal proximity to childbirth is associated with features of breast cancer associated with poor outcomes, including tumor stage, estrogen receptor (ER) status, and risk for distant metastasis and breast cancer-specific mortality, using a population database from the state of Utah. DESIGN, SETTING, AND PARTICIPANTS This population-based cohort study using the Utah Population Database (UPDB) included individuals with stage I to III breast cancer diagnosed at age 45 years or younger between 1996 and 2017, followed-up until February 2020. Participant data were analyzed from November 2019 to August 2022. EXPOSURE The primary exposures were no prior childbirth or time between most recent childbirth and breast cancer diagnosis. Patients were grouped by diagnoses within less than 5 years, 5 to less than 10 years, or 10 years or more since recent childbirth. MAIN OUTCOMES AND MEASURES The 2 primary outcomes were distant metastasis-free survival and breast cancer-specific death. Cox proportional hazard models were used to investigate associations between exposures and outcomes adjusting for diagnosis year, patient age, tumor stage, and estrogen receptor (ER) status. RESULTS Of 2970 individuals with breast cancer diagnosed at age 45 years or younger (mean [SD] age, 39.3 [5.0] years; 12 Black individuals [0.4%], 2679 White individuals [90.2%]), breast cancer diagnosis within 5 years of recent childbirth was independently associated with approximately 1.5-fold elevated risk for metastasis (hazard ratio [HR], 1.5; 95% CI, 1.2-2.0) and breast cancer-specific death (HR, 1.5; 95% CI, 1.1-2.1) compared with nulliparous individuals. For cancers classically considered to have tumor features associated with good outcomes (ie, stage I or II and ER-positive), a postpartum diagnosis was a dominant feature associated with increased risk for metastasis and death (eg, for individuals with ER-positive disease diagnosed within <5 years of childbirth: age-adjusted metastasis HR, 1.5; 95% CI, 1.1-2.1; P = .01; age-adjusted death HR, 1.5; 95% CI, 1.0-2.1; P = .04) compared with nulliparous individuals. Furthermore, liver metastases were specifically increased in the group with diagnosis within 5 years postpartum and with positive ER expression (38 of 83 patients [45.8%]) compared with the nulliparous (28 of 77 patients [36.4%]), although the difference was not statistically significant. Overall, these data implicate parity-associated breast and liver biology in the observed poor outcomes of PPBC. CONCLUSIONS AND RELEVANCE In this cohort study of individuals with breast cancer diagnosed at age 45 years or younger, a postpartum breast cancer diagnosis was a risk factor associated with poor outcomes. Irrespective of ER status, clinical consideration of time between most recent childbirth and breast cancer diagnosis could increase accuracy of prognosis in patients with young-onset breast cancer.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- Division of Oncological Sciences, Oregon Health & Science University, Portland
- Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Solange Bassale
- Biostatistics Shared Resources, Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Sonali Jindal
- Knight Cancer Institute, Oregon Health & Science University, Portland
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland
| | - Alison Fraser
- Pedigree and Population Resource, Population Sciences, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Emily Guinto
- Pedigree and Population Resource, Population Sciences, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Weston Anderson
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland
| | - Motomi Mori
- Department of Biostatistics, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Ken R. Smith
- Pedigree and Population Resource, Population Sciences, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Pepper Schedin
- Knight Cancer Institute, Oregon Health & Science University, Portland
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland
| |
Collapse
|
5
|
Murrow LM, Weber RJ, Caruso JA, McGinnis CS, Phong K, Gascard P, Rabadam G, Borowsky AD, Desai TA, Thomson M, Tlsty T, Gartner ZJ. Mapping hormone-regulated cell-cell interaction networks in the human breast at single-cell resolution. Cell Syst 2022; 13:644-664.e8. [PMID: 35863345 PMCID: PMC9590200 DOI: 10.1016/j.cels.2022.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/02/2022] [Accepted: 06/22/2022] [Indexed: 01/26/2023]
Abstract
The rise and fall of estrogen and progesterone across menstrual cycles and during pregnancy regulates breast development and modifies cancer risk. How these hormones impact each cell type in the breast remains poorly understood because they act indirectly through paracrine networks. Using single-cell analysis of premenopausal breast tissue, we reveal a network of coordinated transcriptional programs representing the tissue-level response to changing hormone levels. Our computational approach, DECIPHER-seq, leverages person-to-person variability in breast composition and cell state to uncover programs that co-vary across individuals. We use differences in cell-type proportions to infer a subset of programs that arise from direct cell-cell interactions regulated by hormones. Further, we demonstrate that prior pregnancy and obesity modify hormone responsiveness through distinct mechanisms: obesity reduces the proportion of hormone-responsive cells, whereas pregnancy dampens the direct response of these cells to hormones. Together, these results provide a comprehensive map of the cycling human breast.
Collapse
Affiliation(s)
- Lyndsay M Murrow
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Robert J Weber
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Medical Scientist Training Program (MSTP), University of California, San Francisco, San Francisco, CA 94518, USA; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Joseph A Caruso
- Department of Pathology and Helen Diller Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christopher S McGinnis
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kiet Phong
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Philippe Gascard
- Department of Pathology and Helen Diller Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gabrielle Rabadam
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alexander D Borowsky
- Center for Immunology and Infectious Diseases, Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, CA 95696, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Thea Tlsty
- Department of Pathology and Helen Diller Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
6
|
Macrophage C/EBPδ Drives Gemcitabine, but Not 5-FU or Paclitaxel, Resistance of Pancreatic Cancer Cells in a Deoxycytidine-Dependent Manner. Biomedicines 2022; 10:biomedicines10020219. [PMID: 35203429 PMCID: PMC8869168 DOI: 10.3390/biomedicines10020219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 02/04/2023] Open
Abstract
Treatment of pancreatic ductal adenocarcinoma (PDAC), a dismal disease with poor survival rates, is hampered by the high prevalence of chemotherapy resistance. Resistance is accompanied by macrophage infiltration into the tumor microenvironment, and infiltrated macrophages are key players in chemotherapy resistance. In the current manuscript, we identify CCAAT/enhancer-binding protein delta (C/EBPδ) as an important transcription factor driving macrophage-dependent gemcitabine resistance. We show that conditioned medium obtained from wild type macrophages largely diminishes gemcitabine-induced cytotoxicity of PDAC cells, whereas conditioned medium obtained from C/EBPδ-deficient macrophages only minimally affects gemcitabine-induced PDAC cell death. Subsequent analysis of RNA-Seq data identified the pyrimidine metabolism pathway amongst the most significant pathways down-regulated in C/EBPδ-deficient macrophages and size filtration experiments indeed showed that the low molecular weight and free metabolite fraction most effectively induced gemcitabine resistance. In line with a role for pyrimidines, we next show that supplementing macrophage conditioned medium with deoxycytidine overruled the effect of macrophage conditioned media on gemcitabine resistance. Consistently, macrophage C/EBPδ-dependent resistance is specific for gemcitabine and does not affect paclitaxel or 5-FU-induced cytotoxicity. Overall, we thus show that C/EBPδ-dependent deoxycytidine biosynthesis in macrophages induces gemcitabine resistance of pancreatic cancer cells.
Collapse
|
7
|
Xu LQ, Yao LJ, Jiang D, Zhou LJ, Chen M, Liao WZ, Zou WH, Peng HJ. A uracil auxotroph Toxoplasma gondii exerting immunomodulation to inhibit breast cancer growth and metastasis. Parasit Vectors 2021; 14:601. [PMID: 34895326 PMCID: PMC8665513 DOI: 10.1186/s13071-021-05032-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Breast cancer is the most common cause of cancer-related death among women, and prognosis is especially poor for patients with triple-negative breast cancer (TNBC); therefore, there is an urgent need for new effective therapies. Recent studies have demonstrated that the uracil auxotroph Toxoplasma gondii vaccine displays anti-tumor effects. Here, we examined the immunotherapy effects of an attenuated uracil auxotroph strain of T. gondii against 4T1 murine breast cancer. METHODS We constructed a uracil auxotroph T. gondii RH strain via orotidine 5'-monophosphate decarboxylase gene deletion (RH-Δompdc) with CRISPR/Cas9 technology. The strain's virulence in the T. gondii-infected mice was determined in vitro and in vivo by parasite replication assay, plaque assay, parasite burden detection in mice peritoneal fluids and survival analysis. The immunomodulation ability of the strain was evaluated by cytokine detection. Its anti-tumor effect was evaluated after its in situ inoculation into 4T1 tumors in a mouse model; the tumor volume was measured, and the 4T1 lung metastasis was detected by hematoxylin and eosin and Ki67 antibody staining, and the cytokine levels were measured by an enzyme-linked immunosorbent assay. RESULTS The RH-Δompdc strain proliferated normally when supplemented with uracil, but it was unable to propagate without the addition of uracil and in vivo, which suggested that it was avirulent to the hosts. This mutant showed vaccine characteristics that could induce intense immune responses both in vitro and in vivo by significantly boosting the expression of inflammatory cytokines. Inoculation of RH-Δompdc in situ into the 4T1 tumor inhibited tumor growth, reduced lung metastasis, promoted the survival of the tumor-bearing mice and increased the secretion of Th1 cytokines, including interleukin-12 (IL-12) and interferon-γ (INF-δ), in both the serum and tumor microenvironment (TME). CONCLUSION Inoculation of the uracil auxotroph RH-Δompdc directly into the 4T1 tumor stimulated anti-infection and anti-tumor immunity in mice, and resulted in inhibition of tumor growth and metastasis, promotion of the survival of the tumor-bearing mice and increased secretion of IL-12 and IFN-γ in both the serum and TME. Our findings suggest that the immunomodulation caused by RH-Δompdc could be a potential anti-tumor strategy.
Collapse
Affiliation(s)
- Li-Qing Xu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Li-Jie Yao
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Dan Jiang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Li-Juan Zhou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Min Chen
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Wen-Zhong Liao
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Wei-Hao Zou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Hong-Juan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China.
| |
Collapse
|
8
|
Postpartum breast cancer has a distinct molecular profile that predicts poor outcomes. Nat Commun 2021; 12:6341. [PMID: 34732713 PMCID: PMC8566602 DOI: 10.1038/s41467-021-26505-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/06/2021] [Indexed: 12/21/2022] Open
Abstract
Young women's breast cancer (YWBC) has poor prognosis and known interactions with parity. Women diagnosed within 5-10 years of childbirth, defined as postpartum breast cancer (PPBC), have poorer prognosis compared to age, stage, and biologic subtype-matched nulliparous patients. Genomic differences that explain this poor prognosis remain unknown. In this study, using RNA expression data from clinically matched estrogen receptor positive (ER+) cases (n = 16), we observe that ER+ YWBC can be differentiated based on a postpartum or nulliparous diagnosis. The gene expression signatures of PPBC are consistent with increased cell cycle, T-cell activation and reduced estrogen receptor and TP53 signaling. When applied to a large YWBC cohort, these signatures for ER+ PPBC associate with significantly reduced 15-year survival rates in high compared to low expressing cases. Cumulatively these results provide evidence that PPBC is a unique entity within YWBC with poor prognostic phenotypes.
Collapse
|
9
|
Non-Tumor CCAAT/Enhancer-Binding Protein Delta Potentiates Tumor Cell Extravasation and Pancreatic Cancer Metastasis Formation. Biomolecules 2021; 11:biom11081079. [PMID: 34439745 PMCID: PMC8391339 DOI: 10.3390/biom11081079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/16/2022] Open
Abstract
CCAAT/enhancer-binding protein delta (C/EBPδ) is a transcription factor involved in apoptosis and proliferation, which is downregulated in pancreatic ductal adenocarcinoma (PDAC) cells. Loss of nuclear C/EBPδ in PDAC cells is associated with decreased patient survival and pro-tumorigenic properties in vitro. Interestingly however, next to C/EBPδ expression in tumor cells, C/EBPδ is also expressed by cells constituting the tumor microenvironment and by cells comprising the organs and parenchyma. However, the functional relevance of systemic C/EBPδ in carcinogenesis remains elusive. Here, we consequently assessed the potential importance of C/EBPδ in somatic tissues by utilizing an orthotopic pancreatic cancer model. In doing so, we show that genetic ablation of C/EBPδ does not significantly affect primary tumor growth but has a strong impact on metastases; wildtype mice developed metastases at multiple sites, whilst this was not the case in C/EBPδ-/- mice. In line with reduced metastasis formation in C/EBPδ-/- mice, C/EBPδ-deficiency also limited tumor cell dissemination in a specific extravasation model. Tumor cell extravasation was dependent on the platelet-activating factor receptor (PAFR) as a PAFR antagonist inhibited tumor cell extravasation in wildtype mice but not in C/EBPδ-/- mice. Overall, we show that systemic C/EBPδ facilitates pancreatic cancer metastasis, and we suggest this is due to C/EBPδ-PAFR-dependent tumor cell extravasation.
Collapse
|
10
|
Lefrère H, Lenaerts L, Borges VF, Schedin P, Neven P, Amant F. Postpartum breast cancer: mechanisms underlying its worse prognosis, treatment implications, and fertility preservation. Int J Gynecol Cancer 2021; 31:412-422. [PMID: 33649008 PMCID: PMC7925817 DOI: 10.1136/ijgc-2020-002072] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022] Open
Abstract
Breast cancers that occur in young women up to 5 to 10 years' postpartum are associated with an increased risk for metastasis and death compared with breast cancers diagnosed in young, premenopausal women during or outside pregnancy. Given the trend to delay childbearing, this frequency is expected to increase. The (immuno)biology of postpartum breast cancer is poorly understood and, hence, it is unknown why postpartum breast cancer has an enhanced risk for metastasis or how it should be effectively targeted for improved survival. The poorer prognosis of women diagnosed within 10 years of a completed pregnancy is most often contributed to the effects of mammary gland involution. We will discuss the most recent data and mechanistic insights of the most important processes associated with involution and their role in the adverse effects of a postpartum diagnosis. We will also look into the effect of lactation on breast cancer outcome after diagnosis. In addition, we will discuss the available treatment strategies that are currently being used to treat postpartum breast cancer, keeping in mind the importance of fertility preservation in this group of young women. These additional insights might offer potential therapeutic options for the improved treatment of women with this specific condition.
Collapse
Affiliation(s)
- Hanne Lefrère
- Department of Oncology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Flanders, Belgium
- Department of Gynecology, AVL NKI, Amsterdam, Noord-Holland, The Netherlands
| | - Liesbeth Lenaerts
- Department of Oncology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Flanders, Belgium
| | - Virginia F Borges
- Department of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - Pepper Schedin
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, Colorado, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Patrick Neven
- Department of Oncology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Flanders, Belgium
- Department of Gynecology and Obstetrics, Katholieke Universiteit Leuven UZ Leuven, Leuven, Flanders, Belgium
- Multidisciplinary Breast Centre, UZ-KU Leuven Cancer Institute (LKI), Katholieke Universiteit Leuven UZ Leuven, Leuven, Flanders, Belgium
| | - Frédéric Amant
- Department of Oncology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Flanders, Belgium
- Department of Gynecology, AVL NKI, Amsterdam, Noord-Holland, The Netherlands
- Department of Gynecology and Obstetrics, Katholieke Universiteit Leuven UZ Leuven, Leuven, Flanders, Belgium
- Department of Gynecological Oncology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Ferrucci V, Asadzadeh F, Collina F, Siciliano R, Boccia A, Marrone L, Spano D, Carotenuto M, Chiarolla CM, De Martino D, De Vita G, Macrì A, Dassi L, Vandenbussche J, Marino N, Cantile M, Paolella G, D'Andrea F, di Bonito M, Gevaert K, Zollo M. Prune-1 drives polarization of tumor-associated macrophages (TAMs) within the lung metastatic niche in triple-negative breast cancer. iScience 2020; 24:101938. [PMID: 33426510 PMCID: PMC7779777 DOI: 10.1016/j.isci.2020.101938] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/22/2020] [Accepted: 12/09/2020] [Indexed: 12/16/2022] Open
Abstract
M2-tumor-associated macrophages (M2-TAMs) in the tumor microenvironment represent a prognostic indicator for poor outcome in triple-negative breast cancer (TNBC). Here we show that Prune-1 overexpression in human TNBC patients has positive correlation to lung metastasis and infiltrating M2-TAMs. Thus, we demonstrate that Prune-1 promotes lung metastasis in a genetically engineered mouse model of metastatic TNBC augmenting M2-polarization of TAMs within the tumor microenvironment. Thus, this occurs through TGF-β enhancement, IL-17F secretion, and extracellular vesicle protein content modulation. We also find murine inactivating gene variants in human TNBC patient cohorts that are involved in activation of the innate immune response, cell adhesion, apoptotic pathways, and DNA repair. Altogether, we indicate that the overexpression of Prune-1, IL-10, COL4A1, ILR1, and PDGFB, together with inactivating mutations of PDE9A, CD244, Sirpb1b, SV140, Iqca1, and PIP5K1B genes, might represent a route of metastatic lung dissemination that need future prognostic validations. Prune-1 correlates to M2-TAMs confirming lung metastatic dissemination in GEMM Cytokines and EV proteins are responsible of M2-TAMs polarization processes A small molecule with immunomodulatory properties ameliorates metastatic dissemination Identification of gene variants within immune response and cell adhesion in TNBC
Collapse
Affiliation(s)
- Veronica Ferrucci
- CEINGE, Biotecnologie Avanzate, Naples 80145, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples 80134, Italy.,European School of Molecular Medicine (SEMM), University of Milan, Milan, Italy
| | - Fatemeh Asadzadeh
- CEINGE, Biotecnologie Avanzate, Naples 80145, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples 80134, Italy
| | - Francesca Collina
- Pathology Unit, Istituto Nazionale Tumori-IRCS- Fondazione G.Pascale, Naples 80131, Italy
| | | | | | - Laura Marrone
- CEINGE, Biotecnologie Avanzate, Naples 80145, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples 80134, Italy
| | | | - Marianeve Carotenuto
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples 80134, Italy
| | | | - Daniela De Martino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples 80134, Italy
| | - Gennaro De Vita
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples 80134, Italy
| | | | - Luisa Dassi
- CEINGE, Biotecnologie Avanzate, Naples 80145, Italy
| | - Jonathan Vandenbussche
- VIB-UGent Centre for Medical Biotechnology, Ghent 9052, Belgium.,Department of Biomolecular Medicine, Ghent University, B9052 Ghent, Belgium
| | - Natascia Marino
- CEINGE, Biotecnologie Avanzate, Naples 80145, Italy.,Department of Medicine, Indiana University-Purdue University Indianapolis, Indianapolis 46202, USA
| | - Monica Cantile
- Pathology Unit, Istituto Nazionale Tumori-IRCS- Fondazione G.Pascale, Naples 80131, Italy
| | | | - Francesco D'Andrea
- Dipartimento di Sanità pubblica - AOU, Università; degli Studi di Napoli Federico II, Naples 80131, Italy
| | - Maurizio di Bonito
- Pathology Unit, Istituto Nazionale Tumori-IRCS- Fondazione G.Pascale, Naples 80131, Italy
| | - Kris Gevaert
- VIB-UGent Centre for Medical Biotechnology, Ghent 9052, Belgium.,Department of Biomolecular Medicine, Ghent University, B9052 Ghent, Belgium
| | - Massimo Zollo
- CEINGE, Biotecnologie Avanzate, Naples 80145, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), 'Federico II' University of Naples, Naples 80134, Italy.,European School of Molecular Medicine (SEMM), University of Milan, Milan, Italy.,DAI Medicina di Laboratorio e Trasfusionale, AOU Federico II, Naples 80131, Italy
| |
Collapse
|
12
|
Ambrosone CB, Higgins MJ. Relationships between Breast Feeding and Breast Cancer Subtypes: Lessons Learned from Studies in Humans and in Mice. Cancer Res 2020; 80:4871-4877. [PMID: 32816853 DOI: 10.1158/0008-5472.can-20-0077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/21/2020] [Accepted: 08/06/2020] [Indexed: 12/15/2022]
Abstract
There are differential risk relationships between parity and breast cancer according to estrogen receptor (ER) status, with an increased risk of ER- disease reduced by breastfeeding. This may be particularly relevant for understanding the higher incidence of ER- tumors in Black women, who are more likely to be parous and less likely to breastfeed than other U.S. groups. Potential mechanisms for these relationships may include effects of disordered breast involution on inflammatory milieu in the breast as well as epigenetic reprogramming in the mammary gland, which can affect cell fate decisions in progenitor cell pools. In normal breast tissue, parity has been associated with hypermethylation of FOXA1, a pioneer transcription factor that promotes the luminal phenotype in luminal progenitors, while repressing the basal phenotype. In breast tumors, relationships between FOXA1 methylation and parity were strongest among women who did not breastfeed. Here, we summarize the epidemiologic literature regarding parity, breastfeeding, and breast cancer subtypes, and review potential mechanisms whereby these factors may influence breast carcinogenesis, with a focus on effects on progenitor cell pools in the mammary gland.
Collapse
Affiliation(s)
- Christine B Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
| | - Michael J Higgins
- Department of Cellular and Molecular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
13
|
Farmaki E, Kaza V, Chatzistamou I, Kiaris H. CCL8 Promotes Postpartum Breast Cancer by Recruiting M2 Macrophages. iScience 2020; 23:101217. [PMID: 32535027 PMCID: PMC7300153 DOI: 10.1016/j.isci.2020.101217] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 04/27/2020] [Accepted: 05/26/2020] [Indexed: 12/23/2022] Open
Abstract
The microenvironment of postpartum mammary gland promotes tumor growth and metastasis in animal models and is linked to increased risk of breast cancer and poor disease outcome in patients. Our previous studies showed the involvement of the chemokine CCL8 in breast cancer metastasis through modulation of the tumor-promoting activity of the tumor microenvironment. Here we show that CCL8 is highly expressed during mammary gland involution and enhances the infiltration of M2 subtype macrophages at the second phase of involution. Cancer cell inoculation studies in Ccl8-deficient animals indicate that CCL8 accelerates tumor onset during involution but not in nulliparous animals. Depletion of macrophages abolished the tumor-promoting effect of CCL8 in involution suggesting the specific role of CCL8 in promoting tumor growth by recruiting macrophages. These results underscore the role of CCL8 in the development of postpartum breast cancer and suggest the potential value of targeting CCL8 in disease management. CCL8 exhibits increased expression during mammary gland involution CCL8 has tumor promoting activity and promotes postpartum breast cancer Targeting CCL8 could have beneficial value for the management of postpartum breast cancer
Collapse
Affiliation(s)
- Elena Farmaki
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Vimala Kaza
- Peromyscus Genetic Stock Center, University of South Carolina, Columbia, SC 29208, USA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology and Immunology, University of South Carolina, School of Medicine, Columbia, SC 29209, USA
| | - Hippokratis Kiaris
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA; Peromyscus Genetic Stock Center, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
14
|
Borges VF, Lyons TR, Germain D, Schedin P. Postpartum Involution and Cancer: An Opportunity for Targeted Breast Cancer Prevention and Treatments? Cancer Res 2020; 80:1790-1798. [PMID: 32075799 PMCID: PMC8285071 DOI: 10.1158/0008-5472.can-19-3448] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/24/2020] [Accepted: 02/12/2020] [Indexed: 12/24/2022]
Abstract
Childbirth at any age confers a transient increased risk for breast cancer in the first decade postpartum and this window of adverse effect extends over two decades in women with late-age first childbirth (>35 years of age). Crossover to the protective effect of pregnancy is dependent on age at first pregnancy, with young mothers receiving the most benefit. Furthermore, breast cancer diagnosis during the 5- to 10-year postpartum window associates with high risk for subsequent metastatic disease. Notably, lactation has been shown to be protective against breast cancer incidence overall, with varying degrees of protection by race, multiparity, and lifetime duration of lactation. An effect for lactation on breast cancer outcome after diagnosis has not been described. We discuss the most recent data and mechanistic insights underlying these epidemiologic findings. Postpartum involution of the breast has been identified as a key mediator of the increased risk for metastasis in women diagnosed within 5-10 years of a completed pregnancy. During breast involution, immune avoidance, increased lymphatic network, extracellular matrix remodeling, and increased seeding to the liver and lymph node work as interconnected pathways, leading to the adverse effect of a postpartum diagnosis. We al discuss a novel mechanism underlying the protective effect of breastfeeding. Collectively, these mechanistic insights offer potential therapeutic avenues for the prevention and/or improved treatment of postpartum breast cancer.
Collapse
Affiliation(s)
- Virginia F Borges
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, Colorado.
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Traci R Lyons
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, Colorado
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Doris Germain
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pepper Schedin
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, Colorado.
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
15
|
Regulation of Immunity in Breast Cancer. Cancers (Basel) 2019; 11:cancers11081080. [PMID: 31366131 PMCID: PMC6721298 DOI: 10.3390/cancers11081080] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer affects millions of women worldwide, leading to many deaths and significant economic burden. Although there are numerous treatment options available, the huge potentials of immunotherapy in the management of localized and metastatic breast cancer is currently being explored. However, there are significant gaps in understanding the complex interactions between the immune system and breast cancer. The immune system can be pro-tumorigenic and anti-tumorigenic depending on the cells involved and the conditions of the tumor microenvironment. In this review, we discuss current knowledge of breast cancer, including treatment options. We also give a brief overview of the immune system and comprehensively highlight the roles of different cells of the immune system in breast tumorigenesis, including recent research discoveries. Lastly, we discuss some immunotherapeutic strategies for the management of breast cancer.
Collapse
|
16
|
Tamburini BAJ, Elder AM, Finlon JM, Winter AB, Wessells VM, Borges VF, Lyons TR. PD-1 Blockade During Post-partum Involution Reactivates the Anti-tumor Response and Reduces Lymphatic Vessel Density. Front Immunol 2019; 10:1313. [PMID: 31244852 PMCID: PMC6579890 DOI: 10.3389/fimmu.2019.01313] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/23/2019] [Indexed: 12/18/2022] Open
Abstract
Post-partum breast cancer patients, or breast cancer patients diagnosed within 10 years of last childbirth, are ~3-5 times more likely to develop metastasis in comparison to non-post-partum, or nulliparous, patients. Additionally, post-partum patients have increased tumor-associated lymphatic vessels and LN involvement, including when controlled for size of the primary tumor. In pre-clinical, immune-competent, mouse mammary tumor models of post-partum breast cancer (PPBC), tumor growth and lymphogenous tumor cell spread occur more rapidly in post-partum hosts. Here we report on PD-L1 expression by lymphatic endothelial cells and CD11b+ cells in the microenvironment of post-partum tumors, which is accompanied by an increase in PD-1 expression by T cells. Additionally, we observed increases in PD-L1 and PD-1 in whole mammary tissues during post-partum mammary gland involution; a known driver of post-partum tumor growth, invasion, and metastasis in pre-clinical models. Importantly, implantation of murine mammary tumor cells during post-partum mammary gland involution elicits a CD8+ T cell population that expresses both the co-inhibitory receptors PD-1 and Lag-3. However, upon anti-PD-1 treatment, during post-partum mammary gland involution, the involution-initiated promotional effects on tumor growth are reversed and the PD-1, Lag-3 double positive population disappears. Consequently, we observed an expansion of poly-functional CD8+ T cells that produced both IFNγ and TNFα. Finally, lymphatic vessel frequency decreased significantly following anti-PD-1 suggesting that anti-PD-1/PD-L1 targeted therapies may have efficacy in reducing tumor growth and dissemination in post-partum breast cancer patients.
Collapse
Affiliation(s)
- Beth A Jirón Tamburini
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States.,Department of Immunology and Microbiology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Alan M Elder
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States.,Young Women's' Breast Cancer Translational Program and University of Colorado Cancer Center, Aurora, CO, United States
| | - Jeffrey M Finlon
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Andrew B Winter
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Veronica M Wessells
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States.,Young Women's' Breast Cancer Translational Program and University of Colorado Cancer Center, Aurora, CO, United States
| | - Virginia F Borges
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States.,Young Women's' Breast Cancer Translational Program and University of Colorado Cancer Center, Aurora, CO, United States
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States.,Young Women's' Breast Cancer Translational Program and University of Colorado Cancer Center, Aurora, CO, United States
| |
Collapse
|
17
|
Choudhary RK, Choudhary S, Mukhopadhyay CS, Pathak D, Verma R. Deciphering the transcriptome of prepubertal buffalo mammary glands using RNA sequencing. Funct Integr Genomics 2019; 19:349-362. [PMID: 30467802 DOI: 10.1007/s10142-018-0645-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 10/28/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
Abstract
Although water buffaloes are the main milk-producing animals in Indian subcontinent, only limited attempts have been made to identify canonical pathways and gene regulatory networks operating within the mammary glands of these animals. Such information is important for identifying unique transcriptome signatures in the mammary glands of diseased animals. In this report, we analyzed the transcription profile of 3 prepubertal buffalo mammary glands and identified common genes (mean FPKM > 0.2 in all samples) operating in the glands. Among 19,994 protein coding genes, 14,678 genes expressed and 5316 unique genes did not express in prepubertal buffalo mammary glands. Of these 14,678 expressed genes, 79% comprised a ubiquitous transcriptome that was dominated by very lowly expressed genes (51%). The percentage of rarely, moderately, and abundantly expressed genes was 25%, 2%, and 1%, respectively. Gene Ontology (GO) terms reflected in the expression of common genes (mean FPKM > 5.0) for molecular function were related to binding and catalytic activity. Products of these genes were involved in metabolic and cellular processes and belong to nucleic acid binding proteins. The canonical pathways for growth of mammary glands included integrin signaling, inflammation, GnRH and Wnt pathways. KEGG enriched pathways revealed many pathways of cancer including ribosome, splisosome, endocytosis, and ubiquitin-mediated proteolysis, pathways for viral infection, and bacterial invasion of epithelial. Highly expressed genes (mean FPKM > 500 included beta-actin (ACTB), beta-2 microglobulin (B2M), caseins (CSN2, CNS3), collagens (COL1A1, COL3A1), translation elongation factors (EEF1A1, EEF1G, EEF2), keratins (KRT15, KRT19), major histocompatibility complex genes (CD74, JSP.1), vimentin (VIM), and osteopontin (SPP1). Interestingly, expression of milk protein genes in prepubertal glands opens possible roles of these genes in development of mammary glands. We report the whole transcriptomic signature of prepubertal buffalo mammary gland and indicated its molecular signature is similar to cancer type.
Collapse
Affiliation(s)
- Ratan K Choudhary
- School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, 141004, India.
- Department of Animal and Veterinary Sciences, University of Vermont, Burlington, VT, 05405, USA.
| | - Shanti Choudhary
- School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, 141004, India
| | - C S Mukhopadhyay
- School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, 141004, India
| | - Devendra Pathak
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Science University, Ludhiana, Punjab, 141004, India
| | - Ramneek Verma
- School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, 141004, India
| |
Collapse
|
18
|
Autophagy and unfolded protein response (UPR) regulate mammary gland involution by restraining apoptosis-driven irreversible changes. Cell Death Discov 2018; 4:40. [PMID: 30345078 PMCID: PMC6186758 DOI: 10.1038/s41420-018-0105-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 08/01/2018] [Accepted: 09/05/2018] [Indexed: 12/31/2022] Open
Abstract
The postnatal mammary gland undergoes repeated cycles of proliferation and cell death, most notably when the fully differentiated (lactating) gland dedifferentiates to a prelactation state. Accumulation of milk proteins in the secretory epithelium creates the stress signal that triggers this process (involution). How this stress is perceived, and the cellular processes that are subsequently activated, remain unclear. We now report that Unfolded Protein Response (UPR), autophagy, and apoptosis related genes cluster separately during lactation and involution in the mouse mammary gland. Time-course experiments in rodents show that autophagy and UPR signaling are tightly co-regulated at the transition from reversible to irreversible involution. Inhibition of autophagy by chloroquine or genetic deletion of one ATG7 allele enhanced progression of mammary involution into the irreversible phase, as characterized by an early/precocious induction of apoptosis. These are the first preclinical in vivo data in support of a clinical trial testing an autophagy inhibitor for prevention of intraductal breast malignancy progression to invasive breast cancer. In marked contrast, stimulation of autophagy by low dose tunicamycin treatment reduced apoptosis and extended the reversible phase of involution by sustaining the secretory epithelium. Autophagy stimulators could be used short-term to promote lactation in women experiencing difficulties or irregularities in nursing. Taken together, these data indicate that UPR and autophagy play a key role in regulating the balance between cell survival and apoptosis during normal mammary gland regression.
Collapse
|
19
|
Reddy JP, Atkinson RL, Larson R, Burks JK, Smith D, Debeb BG, Ruffell B, Creighton CJ, Bambhroliya A, Reuben JM, Van Laere SJ, Krishnamurthy S, Symmans WF, Brewster AM, Woodward WA. Mammary stem cell and macrophage markers are enriched in normal tissue adjacent to inflammatory breast cancer. Breast Cancer Res Treat 2018; 171:283-293. [PMID: 29858753 DOI: 10.1007/s10549-018-4835-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
Abstract
INTRODUCTION We hypothesized that breast tissue not involved by tumor in inflammatory breast cancer (IBC) patients contains intrinsic differences, including increased mammary stem cells and macrophage infiltration, which may promote the IBC phenotype. MATERIALS AND METHODS Normal breast parenchyma ≥ 5 cm away from primary tumors was obtained from mastectomy specimens. This included an initial cohort of 8 IBC patients and 60 non-IBC patients followed by a validation cohort of 19 IBC patients and 25 non-IBC patients. Samples were immunostained for either CD44+CD49f+CD133/2+ mammary stem cell markers or the CD68 macrophage marker and correlated with IBC status. Quantitation of positive cells was determined using inForm software from PerkinElmer. We also examined the association between IBC status and previously published tumorigenic stem cell and IBC tumor signatures in the validation cohort samples. RESULTS 8 of 8 IBC samples expressed isolated CD44+CD49f+CD133/2+ stem cell marked cells in the initial cohort as opposed to 0/60 non-IBC samples (p = 0.001). Similarly, the median number of CD44+CD49f+CD133/2+ cells was significantly higher in the IBC validation cohort as opposed to the non-IBC validation cohort (25.7 vs. 14.2, p = 0.007). 7 of 8 IBC samples expressed CD68 + histologically confirmed macrophages in initial cohort as opposed to 12/48 non-IBC samples (p = 0.001). In the validation cohort, the median number of CD68 + cells in IBC was 3.7 versus 1.0 in the non-IBC cohort (p = 0.06). IBC normal tissue was positively associated with a tumorigenic stem cell signature (p = 0.02) and with a 79-gene IBC signature (p < 0.001). CONCLUSIONS Normal tissue from IBC patients is enriched for both mammary stem cells and macrophages and has higher association with both a tumorigenic stem cell signature and IBC-specific tumor signature. Collectively, these data suggest that IBC normal tissue differs from non-IBC tissue. Whether these changes occur before the tumor develops or is induced by tumor warrants further investigation.
Collapse
Affiliation(s)
- Jay P Reddy
- Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rachel L Atkinson
- Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard Larson
- Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared K Burks
- Flow Cytometry and Cell Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel Smith
- Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bisrat G Debeb
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Chad J Creighton
- Department of Medicine and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arvind Bambhroliya
- Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven J Van Laere
- Faculty of Medicine and Health Sciences, Center for Oncological Research, University of Antwerp, Antwerp, Belgium
| | - Savitri Krishnamurthy
- Department of Breast Surgical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William F Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abenaa M Brewster
- Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wendy A Woodward
- Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, Unit 1202, Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
20
|
Gene set analysis of post-lactational mammary gland involution gene signatures in inflammatory and triple-negative breast cancer. PLoS One 2018; 13:e0192689. [PMID: 29617367 PMCID: PMC5884491 DOI: 10.1371/journal.pone.0192689] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 01/29/2018] [Indexed: 12/31/2022] Open
Abstract
Background Epidemiological studies have found that triple-negative breast cancer (TNBC) and TN inflammatory breast cancer (IBC) are associated with lower frequency and duration of breast-feeding compared to non-TNBC and non-TN IBC, respectively. Limited breast-feeding could reflect abrupt or premature involution and contribute to a “primed” stroma that is permissive to the migration of cancer cells typical of IBC. We hypothesized that gene expression related to abrupt mammary gland involution after forced weaning may be enriched in the tissues of IBC patients and, if so, provide a potential correlation between limited breast-feeding and the development of aggressive breast cancer. Methods We utilized the Short Time-series Expression Miner (STEM) program to cluster significant signatures from two independent studies that analyzed gene expression at multiple time-points of mouse mammary gland involution. Using 10 significant signatures, we performed gene ontology analysis and gene set enrichment analysis (GSEA) on training and validation sets from human breast cancer gene expression data to identify specific genes that are enriched in IBC compared to non-IBC and in TN compared to non-TN in IBC and non-IBC groups. Results Examining the combined data, we identified 10 involution gene clusters (Inv1-10) that share time-dependent regulation after forced weaning. Inv5 was the only cluster significantly enriched in IBC in the training and validation set (nominal p-values <0.05) and only by unadjusted p-values (FDR q-values 0.26 and 0.46 respectively). Eight genes in Inv5 are upregulated in both the training and validation sets in IBC. Combining the training and validation sets, both Inv5 and Inv6 have nominal p-values <0.05 and q-values 0.39 and 0.20, respectively. The time course for both clusters includes genes that change within 12 hours after forced weaning. Conclusions Results from this in silico study suggest correlation between molecular events during abrupt involution and aggressive breast cancer. Specifically, candidate genes from Inv5 merit functional investigation regarding the role of limited breast-feeding in IBC development.
Collapse
|
21
|
Dai WT, Zou YX, White RR, Liu JX, Liu HY. Transcriptomic profiles of the bovine mammary gland during lactation and the dry period. Funct Integr Genomics 2018; 18:125-140. [PMID: 29275436 DOI: 10.1007/s10142-017-0580-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/06/2017] [Accepted: 12/08/2017] [Indexed: 01/22/2023]
Abstract
The initiation and maintenance of lactation are complex phenomena governed by biochemical and endocrine processes in the mammary gland (MG). Although DNA-based approaches have been used to study the onset of lactation, more comprehensive RNA-based techniques may be critical in furthering our understanding of gene alterations that occur to support lactation in the bovine MG. To further determine how gene profiles vary during lactation compared with the dry period, RNA-seq transcriptomic analysis was used to identify differentially expressed genes (DEG) in bovine MG tissues from animals that were lactating and not lactating. A total of 881 DEG (605 upregulated and 276 downregulated) were identified in MG of 3 lactating Chinese Holstein dairy cows versus the 3 dry cows. The subcellular analysis showed that the upregulated genes were most abundantly located in "integral to membrane" and "mitochondrion," and the top number of downregulated genes existed in "nucleus" and "cytoplasm." The functional analysis indicated that the DEG were primarily associated with the support of lactation processes. The genes in higher abundance were most related to "metabolic process," "oxidation-reduction process," "transport" and "signal transduction," protein synthesis-related processes (transcription, translation, protein modifications), and some MG growth-associated processes (cell proliferation/cycle/apoptosis). The downregulated genes were mainly involved in immune-related processes (inflammatory/immune/defense responses). The KEGG analysis suggested that protein synthesis-related pathways (such as protein digestion and absorption; protein processing in endoplasmic reticulum; and glycine, serine, and threonine metabolism) were highly and significantly enriched in the bovine MG of lactating cows compared to dry cows. The results suggested that the dry cows had decreased capacity for protein synthesis, energy generation, and cell growth but enhanced immune response. Collectively, this reduced capacity in dry cows supports the physiological demands of the next lactation and the coordinated metabolic changes that occur to support these demands. A total of 51 identified DEG were validated by RT-PCR, and consistent results were found between RT-PCR and the transcriptomic analysis. This work provides a profile of gene-associated changes that occur during lactation and can be used to facilitate further investigation of the mechanisms underlying lactation in dairy cows.
Collapse
Affiliation(s)
- Wen-Ting Dai
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Yi-Xuan Zou
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Robin R White
- Department of Animal and Poultry Science, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Jian-Xin Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Hong-Yun Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| |
Collapse
|
22
|
Li Y, Pang Z, Dong X, Liao X, Deng H, Liao C, Liao Y, Chen G, Huang L. MUC1 induces M2 type macrophage influx during postpartum mammary gland involution and triggers breast cancer. Oncotarget 2017; 9:3446-3458. [PMID: 29423058 PMCID: PMC5790475 DOI: 10.18632/oncotarget.23316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/19/2017] [Indexed: 01/28/2023] Open
Abstract
The microenvironment of postpartum mammary gland involution (PMI) has been linked to the increased risk of breast cancer and poor outcome of patients. Nevertheless the mechanism underlying regulates the microenvironment remains largely unknown. MUC1, which is abnormally overexpressed in most breast cancer, is physiologically expressed in PMI. Using MUC1 cytoplasm domain (MUC1-CD) transgenic mice, we reveal that the overexpression of MUC1-CD in mammary epithelial cells increases M2 type macrophage infiltration in PMI. By sustain activating p50, MUC1 upregulates M2 macrophage chemo-attractants and the anti-apoptotic protein Bcl-xL. Because of the tumor promotional microenvironments and reduced apoptosis, MUC1-CD delays PMI process and results in atypical phenotype in multiparous mice mammary. This finding is further supported by the positive association between the expression of MUC1 and p50 in Luminal A and Luminal B subtypes through analyzing breast cancer databases. Taken together, our study demonstrates that MUC1-CD plays an important role in regulating microenvironment of PMI and promoting postpartum mammary tumorigenicity, providing novel prevention and treatment strategies against postpartum breast cancer.
Collapse
Affiliation(s)
- Yuan Li
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, P.R. China
| | - Zhi Pang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Xinran Dong
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Xiaodong Liao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Huayun Deng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Chunhua Liao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yahui Liao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Guoqiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Lei Huang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
23
|
Platonova NA, Orlov IA, Klotchenko SA, Babich VS, Ilyechova EY, Babich PS, Garmai YP, Vasin AV, Tsymbalenko NV, Puchkova LV. Ceruloplasmin gene expression profile changes in the rat mammary gland during pregnancy, lactation and involution. J Trace Elem Med Biol 2017; 43:126-134. [PMID: 28089327 DOI: 10.1016/j.jtemb.2016.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/31/2016] [Accepted: 12/31/2016] [Indexed: 11/19/2022]
Abstract
Copper metabolism disturbances in mammary gland (MG) cells have severe consequences in newborns. The mechanism that controls the balance of copper in the MG has not been thoroughly characterized. Four primary copper homeostasis genes in mammals: (1) ceruloplasmin (Cp) encoding multifunction multicopper blue (ferr)oxidase; (2) CTR1 encoding high affinity copper importer 1; and (3 and 4) two similar genes encoding Cu(I)/Cu(II)-ATPases P1 type (ATP7A and ATP7B) responsible for copper efflux from the cells and metallation of cuproenzymes formed in the Golgi complex are expressed in MG. This study aimed to characterize expression of these genes during pregnancy, lactation and forced involution in the rat MG. We found that Cp anchored to the plasma membrane and ATP7A were expressed during pregnancy and lactation. Soluble Cp and ATP7B were highly expressed in lactating MG decreasing to its ending. CTR1 activity increased during MG growth and reached its maximum at postpartum and then it decreased until the end of lactation. During early forced MG involution, Cp gene expression persisted; while a form of Cp that lacked exon 18 appeared. We suggest that Cp gene expressional changes at the transcriptional and posttranscriptional level reflect various physiological functions of Cp proteins during MG remodeling.
Collapse
Affiliation(s)
- Natalia A Platonova
- Institute of Experimental Medicine, Pavlova str., 12, St., Petersburg 197376, Russia
| | - Iurii A Orlov
- ITMO University, Kronverksky av., 49, St., Petersburg 197101, Russia; Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya str., 29, St., Petersburg 195251, Russia.
| | - Sergey A Klotchenko
- Institute of Experimental Medicine, Pavlova str., 12, St., Petersburg 197376, Russia
| | - Victor S Babich
- School of Liberal Arts and Sciences, Mercy College of Health Sciences, Des Moines, IA, USA
| | - Ekaterina Y Ilyechova
- Institute of Experimental Medicine, Pavlova str., 12, St., Petersburg 197376, Russia; ITMO University, Kronverksky av., 49, St., Petersburg 197101, Russia
| | - Polina S Babich
- Institute of Experimental Medicine, Pavlova str., 12, St., Petersburg 197376, Russia
| | - Yuri P Garmai
- Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya str., 29, St., Petersburg 195251, Russia
| | - Andrey V Vasin
- Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya str., 29, St., Petersburg 195251, Russia
| | - Nadezhda V Tsymbalenko
- Institute of Experimental Medicine, Pavlova str., 12, St., Petersburg 197376, Russia; ITMO University, Kronverksky av., 49, St., Petersburg 197101, Russia
| | - Liudmila V Puchkova
- Institute of Experimental Medicine, Pavlova str., 12, St., Petersburg 197376, Russia; ITMO University, Kronverksky av., 49, St., Petersburg 197101, Russia; Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya str., 29, St., Petersburg 195251, Russia
| |
Collapse
|
24
|
Law AMK, Lim E, Ormandy CJ, Gallego-Ortega D. The innate and adaptive infiltrating immune systems as targets for breast cancer immunotherapy. Endocr Relat Cancer 2017; 24:R123-R144. [PMID: 28193698 PMCID: PMC5425956 DOI: 10.1530/erc-16-0404] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 02/13/2017] [Indexed: 12/15/2022]
Abstract
A cancer cell-centric view has long dominated the field of cancer biology. Research efforts have focussed on aberrant cancer cell signalling pathways and on changes to cancer cell DNA. Mounting evidence demonstrates that many cancer-associated cell types within the tumour stroma co-evolve and support tumour growth and development, greatly modifying cancer cell behaviour, facilitating invasion and metastasis and controlling dormancy and sensitivity to drug therapy. Thus, these stromal cells represent potential targets for cancer therapy. Among these cell types, immune cells have emerged as a promising target for therapy. The adaptive and the innate immune system play an important role in normal mammary development and breast cancer. The number of infiltrating adaptive immune system cells with tumour-rejecting capacity, primarily, T lymphocytes, is lower in breast cancer compared with other cancer types, but infiltration occurs in a large proportion of cases. There is strong evidence demonstrating the importance of the immunosuppressive role of the innate immune system during breast cancer progression. A consideration of components of both the innate and the adaptive immune system is essential for the design and development of immunotherapies in breast cancer. In this review, we focus on the importance of immunosuppressive myeloid-derived suppressor cells (MDSCs) as potential targets for breast cancer therapy.
Collapse
Affiliation(s)
- Andrew M K Law
- Tumour Development GroupThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Cancer Biology LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Elgene Lim
- Connie Johnson Breast Cancer Research LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Christopher J Ormandy
- Cancer Biology LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - David Gallego-Ortega
- Tumour Development GroupThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| |
Collapse
|
25
|
Guo Q, Minnier J, Burchard J, Chiotti K, Spellman P, Schedin P. Physiologically activated mammary fibroblasts promote postpartum mammary cancer. JCI Insight 2017; 2:e89206. [PMID: 28352652 DOI: 10.1172/jci.insight.89206] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Women diagnosed with breast cancer within 5 years of childbirth have poorer prognosis than nulliparous or pregnant women. Weaning-induced breast involution is implicated, as the collagen-rich, immunosuppressive microenvironment of the involuting mammary gland is tumor promotional in mice. To investigate the role of mammary fibroblasts, isolated mammary PDGFRα+ cells from nulliparous and postweaning mice were assessed for activation phenotype and protumorigenic function. Fibroblast activation during involution was evident by increased expression of fibrillar collagens, lysyl oxidase, Tgfb1, and Cxcl12 genes. The ability of mammary tumors to grow in an isogenic, orthotopic transplant model was increased when tumor cells were coinjected with involution-derived compared with nulliparous-derived mammary fibroblasts. Mammary tumors in the involution-fibroblast group had increased Ly6C+ monocytes at the tumor border, and decreased CD8+ T cell infiltration and tumor cell death. Ibuprofen treatment suppressed involution-fibroblast activation and tumor promotional capacity, concurrent with decreases in tumor Ly6C+ monocytes, and increases in intratumoral CD8+ T cell infiltration, granzyme levels, and tumor cell death. In total, our data identify a COX/prostaglandin E2 (PGE2)-dependent activated mammary fibroblast within the involuting mammary gland that displays protumorigenic, immunosuppressive activity, identifying fibroblasts as potential targets for the prevention and treatment of postpartum breast cancer.
Collapse
Affiliation(s)
- Qiuchen Guo
- Department of Cell, Developmental and Cancer Biology
| | | | | | - Kami Chiotti
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Paul Spellman
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
26
|
LPA receptor activity is basal specific and coincident with early pregnancy and involution during mammary gland postnatal development. Sci Rep 2016; 6:35810. [PMID: 27808166 PMCID: PMC5093903 DOI: 10.1038/srep35810] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 10/06/2016] [Indexed: 01/08/2023] Open
Abstract
During pregnancy, luminal and basal epithelial cells of the adult mammary gland proliferate and differentiate resulting in remodeling of the adult gland. While pathways that control this process have been characterized in the gland as a whole, the contribution of specific cell subtypes, in particular the basal compartment, remains largely unknown. Basal cells provide structural and contractile support, however they also orchestrate the communication between the stroma and the luminal compartment at all developmental stages. Using RNA-seq, we show that basal cells are extraordinarily transcriptionally dynamic throughout pregnancy when compared to luminal cells. We identified gene expression changes that define specific basal functions acquired during development that led to the identification of novel markers. Enrichment analysis of gene sets from 24 mouse models for breast cancer pinpoint to a potential new function for insulin-like growth factor 1 (Igf1r) in the basal epithelium during lactogenesis. We establish that β-catenin signaling is activated in basal cells during early pregnancy, and demonstrate that this activity is mediated by lysophosphatidic acid receptor 3 (Lpar3). These findings identify novel pathways active during functional maturation of the adult mammary gland.
Collapse
|
27
|
Borges VF, Elder AM, Lyons TR. Deciphering Pro-Lymphangiogenic Programs during Mammary Involution and Postpartum Breast Cancer. Front Oncol 2016; 6:227. [PMID: 27853703 PMCID: PMC5090124 DOI: 10.3389/fonc.2016.00227] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/10/2016] [Indexed: 12/12/2022] Open
Abstract
Postpartum breast cancers are a highly metastatic subset of young women’s breast cancers defined as breast cancers diagnosed in the postpartum period or within 5 years of last child birth. Women diagnosed with postpartum breast cancer are nearly twice as likely to develop metastasis and to die from breast cancer when compared with nulliparous women. Additionally, epidemiological studies utilizing multiple cohorts also suggest that nearly half of all breast cancers in women aged <45 qualify as postpartum cases. Understanding the biology that underlies this increased risk for metastasis and death may lead to identification of targeted interventions that will benefit the large number of young women with breast cancer who fall into this subset. Preclinical mouse models of postpartum breast cancer have revealed that breast tumor cells become more aggressive if they are present during the normal physiologic process of postpartum mammary gland involution in mice. As involution appears to be a period of lymphatic growth and remodeling, and human postpartum breast cancers have high peritumor lymphatic vessel density (LVD) and increased incidence of lymph node metastasis (1, 2), we propose that novel insight into is to be gained through the study of the biological mechanisms driving normal postpartum mammary lymphangiogenesis as well as in the microenvironment of postpartum tumors.
Collapse
Affiliation(s)
- Virginia F Borges
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, CO, USA; Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alan M Elder
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, CO, USA; Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Traci R Lyons
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, CO, USA; Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
28
|
Woodward WA. Inflammatory breast cancer: unique biological and therapeutic considerations. Lancet Oncol 2016; 16:e568-e576. [PMID: 26545845 DOI: 10.1016/s1470-2045(15)00146-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/29/2015] [Accepted: 07/06/2015] [Indexed: 12/29/2022]
Abstract
Through the concerted efforts of many patients, health-care providers, legislators, and other supporters, the past decade has seen the development of the first clinics dedicated to the care of patients with inflammatory breast cancer in the USA and other countries. Together with social networking, advocacy, and education, a few specialised centres have had substantial increases in patient numbers (in some cases ten times higher), which has further expanded the community of science and advocacy and increased the understanding of the disease process. Although inflammatory breast cancer is considered rare, constituting only 2-4% of breast cancer cases, poor prognosis means that patients with the disease account for roughly 10% of breast cancer mortality annually in the USA. I propose that the unique presentation of inflammatory breast cancer might require specific, identifiable changes in the breast parenchyma that occur before the tumour-initiating event. This would make the breast tissue itself a tumour-promoting medium that should be treated as a component of the pathology in multidisciplinary treatment and should be further studied for complementary targets to inhibit the pathobiology that is specific to inflammatory breast cancer.
Collapse
Affiliation(s)
- Wendy A Woodward
- Department of Radiation Oncology and MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
29
|
Curran CS, Carrillo ER, Ponik SM, Keely PJ. Collagen density regulates xenobiotic and hypoxic response of mammary epithelial cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:114-124. [PMID: 25481308 PMCID: PMC4323890 DOI: 10.1016/j.etap.2014.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 10/27/2014] [Indexed: 06/04/2023]
Abstract
Breast density, where collagen I is the dominant component, is a significant breast cancer risk factor. Cell surface integrins interact with collagen, activate focal adhesion kinase (FAK), and downstream cell signals associated with xenobiotics (AhR, ARNT) and hypoxia (HIF-1α, ARNT). We examined if mammary cells cultured in high density (HD) or low density (LD) collagen gels affected xenobiotic or hypoxic responses. ARNT production was significantly reduced by HD culture and in response to a FAK inhibitor. Consistent with a decrease in ARNT, AhR and HIF-1α reporter activation and VEGF production was lower in HD compared to LD. However, P450 production was enhanced in HD and induced by AhR and HIF-1α agonists, possibly in response to increased NF-κB activaton. Thus, collagen density differentially regulates downstream cell signals of AhR and HIF-1α by modulating the activity of FAK, the release of NF-κB transcriptional factors, and the levels of ARNT.
Collapse
Affiliation(s)
- Colleen S Curran
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| | - Esteban R Carrillo
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| | - Patricia J Keely
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, WI 53706, USA.
| |
Collapse
|
30
|
Figueroa JD, Pfeiffer RM, Patel DA, Linville L, Brinton LA, Gierach GL, Yang XR, Papathomas D, Visscher D, Mies C, Degnim AC, Anderson WF, Hewitt S, Khodr ZG, Clare SE, Storniolo AM, Sherman ME. Terminal duct lobular unit involution of the normal breast: implications for breast cancer etiology. J Natl Cancer Inst 2014; 106:dju286. [PMID: 25274491 DOI: 10.1093/jnci/dju286] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Greater degrees of terminal duct lobular unit (TDLU) involution have been linked to lower breast cancer risk; however, factors that influence this process are poorly characterized. METHODS To study this question, we developed three reproducible measures that are inversely associated with TDLU involution: TDLU counts, median TDLU span, and median acini counts/TDLU. We determined factors associated with TDLU involution using normal breast tissues from 1938 participants (1369 premenopausal and 569 postmenopausal) ages 18 to 75 years in the Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center. Multivariable zero-inflated Poisson models were used to estimate relative risks (RRs) and 95% confidence intervals (95% CIs) for factors associated with TDLU counts, and multivariable ordinal logistic regression models were used to estimate odds ratios (ORs) and 95% CIs for factors associated with categories of median TDLU span and acini counts/TDLU. RESULTS All TDLU measures started declining in the third age decade (all measures, two-sided P trend ≤ .001); and all metrics were statistically significantly lower among postmenopausal women. Nulliparous women demonstrated lower TDLU counts compared with uniparous women (among premenopausal women, RR = 0.79, 95% CI = 0.73 to 0.85; among postmenopausal, RR = 0.67, 95% CI = 0.56 to 0.79); however, rates of age-related TDLU decline were faster among parous women. Other factors were related to specific measures of TDLU involution. CONCLUSION Morphometric analysis of TDLU involution warrants further evaluation to understand the pathogenesis of breast cancer and assessing its role as a progression marker for women with benign biopsies or as an intermediate endpoint in prevention studies.
Collapse
Affiliation(s)
- Jonine D Figueroa
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS).
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Deesha A Patel
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Laura Linville
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Gretchen L Gierach
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Xiaohong R Yang
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Daphne Papathomas
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Daniel Visscher
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Carolyn Mies
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Amy C Degnim
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - William F Anderson
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Stephen Hewitt
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Zeina G Khodr
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Susan E Clare
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Anna Maria Storniolo
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| | - Mark E Sherman
- Division of Cancer Epidemiology and Genetics (JDF, RMP, DAP, LL, LAB, GLG, XRY, DP, WFA, ZGK, MES), Laboratory of Pathology (SH), and Division of Cancer Prevention (MES), National Cancer Institute, Bethesda, MD; Mayo Clinic Cancer Center, Rochester, MN (DV, ACD); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA (CM); Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL (SEC); Susan G. Komen Tissue Bank at the Indiana University Simon Cancer Center, Indianapolis, IN (AMS)
| |
Collapse
|
31
|
Stanford JC, Young C, Hicks D, Owens P, Williams A, Vaught DB, Morrison MM, Lim J, Williams M, Brantley-Sieders DM, Balko JM, Tonetti D, Earp HS, Cook RS. Efferocytosis produces a prometastatic landscape during postpartum mammary gland involution. J Clin Invest 2014; 124:4737-52. [PMID: 25250573 DOI: 10.1172/jci76375] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 08/13/2014] [Indexed: 12/27/2022] Open
Abstract
Breast cancers that occur in women 2-5 years postpartum are more frequently diagnosed at metastatic stages and correlate with poorer outcomes compared with breast cancers diagnosed in young, premenopausal women. The molecular mechanisms underlying the malignant severity associated with postpartum breast cancers (ppBCs) are unclear but relate to stromal wound-healing events during postpartum involution, a dynamic process characterized by widespread cell death in milk-producing mammary epithelial cells (MECs). Using both spontaneous and allografted mammary tumors in fully immune-competent mice, we discovered that postpartum involution increases mammary tumor metastasis. Cell death was widespread, not only occurring in MECs but also in tumor epithelium. Dying tumor cells were cleared through receptor tyrosine kinase MerTK-dependent efferocytosis, which robustly induced the transcription of genes encoding wound-healing cytokines, including IL-4, IL-10, IL-13, and TGF-β. Animals lacking MerTK and animals treated with a MerTK inhibitor exhibited impaired efferocytosis in postpartum tumors, a reduction of M2-like macrophages but no change in total macrophage levels, decreased TGF-β expression, and a reduction of postpartum tumor metastasis that was similar to the metastasis frequencies observed in nulliparous mice. Moreover, TGF-β blockade reduced postpartum tumor metastasis. These data suggest that widespread cell death during postpartum involution triggers efferocytosis-induced wound-healing cytokines in the tumor microenvironment that promote metastatic tumor progression.
Collapse
|
32
|
Hyaluronan and RHAMM in wound repair and the "cancerization" of stromal tissues. BIOMED RESEARCH INTERNATIONAL 2014; 2014:103923. [PMID: 25157350 PMCID: PMC4137499 DOI: 10.1155/2014/103923] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 07/04/2014] [Indexed: 12/12/2022]
Abstract
Tumors and wounds share many similarities including loss of tissue architecture, cell polarity and cell differentiation, aberrant extracellular matrix (ECM) remodeling (Ballard et al., 2006) increased inflammation, angiogenesis, and elevated cell migration and proliferation. Whereas these changes are transient in repairing wounds, tumors do not regain tissue architecture but rather their continued progression is fueled in part by loss of normal tissue structure. As a result tumors are often described as wounds that do not heal. The ECM component hyaluronan (HA) and its receptor RHAMM have both been implicated in wound repair and tumor progression. This review highlights the similarities and differences in their roles during these processes and proposes that RHAMM-regulated wound repair functions may contribute to “cancerization” of the tumor microenvironment.
Collapse
|
33
|
Fornetti J, Martinson HA, Betts CB, Lyons TR, Jindal S, Guo Q, Coussens LM, Borges VF, Schedin P. Mammary gland involution as an immunotherapeutic target for postpartum breast cancer. J Mammary Gland Biol Neoplasia 2014; 19:213-28. [PMID: 24952477 PMCID: PMC4363120 DOI: 10.1007/s10911-014-9322-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 06/09/2014] [Indexed: 12/24/2022] Open
Abstract
Postpartum mammary gland involution has been identified as tumor-promotional and is proposed to contribute to the increased rates of metastasis and poor survival observed in postpartum breast cancer patients. In rodent models, the involuting mammary gland microenvironment is sufficient to induce enhanced tumor cell growth, local invasion, and metastasis. Postpartum involution shares many attributes with wound healing, including upregulation of genes involved in immune responsiveness and infiltration of tissue by immune cells. In rodent models, treatment with non-steroidal anti-inflammatory drugs (NSAIDs) ameliorates the tumor-promotional effects of involution, consistent with the immune milieu of the involuting gland contributing to tumor promotion. Currently, immunotherapy is being investigated as a means of breast cancer treatment with the purpose of identifying ways to enhance anti-tumor immune responses. Here we review evidence for postpartum mammary gland involution being a uniquely defined 'hot-spot' of pro-tumorigenic immune cell infiltration, and propose that immunotherapy should be explored for prevention and treatment of breast cancers that arise in this environment.
Collapse
Affiliation(s)
- Jaime Fornetti
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
- Program in Reproductive Sciences, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Holly A. Martinson
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USA
| | - Courtney B. Betts
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
- Cell Biology, Stem cells, and Development, 12801 E 17th Ave, Aurora, CO 80045, USA
| | - Traci R. Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
| | - Sonali Jindal
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
| | - Qiuchen Guo
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USA
| | - Lisa M. Coussens
- Department of Cell & Developmental Biology, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Virginia F. Borges
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
| | - Pepper Schedin
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
- Program in Reproductive Sciences, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USA
- Cell Biology, Stem cells, and Development, 12801 E 17th Ave, Aurora, CO 80045, USA
- Department of Cell & Developmental Biology, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
34
|
Jindal S, Gao D, Bell P, Albrektsen G, Edgerton SM, Ambrosone CB, Thor AD, Borges VF, Schedin P. Postpartum breast involution reveals regression of secretory lobules mediated by tissue-remodeling. Breast Cancer Res 2014; 16:R31. [PMID: 24678808 PMCID: PMC4053254 DOI: 10.1186/bcr3633] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 03/06/2014] [Indexed: 01/05/2023] Open
Abstract
Introduction A postpartum diagnosis of breast cancer is an independent predictor of metastases, however the reason is unknown. In rodents, the window of postpartum mammary gland involution promotes tumor progression, suggesting a role for breast involution in the poor prognosis of human postpartum breast cancers. Rodent mammary gland involution is characterized by the programmed elimination of the secretory lobules laid down in preparation for lactation. This tissue involution process involves massive epithelial cell death, stromal remodeling, and immune cell infiltration with similarities to microenvironments present during wound healing and tumor progression. Here, we characterize breast tissue from premenopausal women with known reproductive histories to determine the extent, duration and cellular mechanisms of postpartum lobular involution in women. Methods Adjacent normal breast tissues from premenopausal women (n = 183) aged 20 to 45 years, grouped by reproductive categories of nulliparous, pregnant and lactating, and by time since last delivery were evaluated histologically and by special stain for lobular area, lobular type composition, apoptosis and immune cell infiltration using computer assisted quantitative methods. Results Human nulliparous glands were composed dominantly of small (approximately 10 acini per lobule) and medium (approximately 35 acini per lobule) sized lobules. With pregnancy and lactation, a >10 fold increase in breast epithelial area was observed compared to nulliparous cases, and lactating glands were dominated by mature lobules (>100 acini per lobule) with secretory morphology. Significant losses in mammary epithelial area and mature lobule phenotypes were observed within 12 months postpartum. By 18 months postpartum, lobular area content and lobule composition were indistinguishable from nulliparous cases, data consistent with postpartum involution facilitating regression of the secretory lobules developed in preparation for lactation. Analyses of apoptosis and immune cell infiltrate confirmed that human postpartum breast involution is characterized by wound healing-like tissue remodeling programs that occur within a narrowed time frame. Conclusions Human postpartum breast involution is a dominant tissue-remodeling process that returns the total lobular area of the gland to a level essentially indistinguishable from the nulliparous gland. Further research is warranted to determine whether the normal physiologic process of postpartum involution contributes to the poor prognosis of postpartum breast cancer.
Collapse
|
35
|
Loss of Igfbp7 causes precocious involution in lactating mouse mammary gland. PLoS One 2014; 9:e87858. [PMID: 24505323 PMCID: PMC3913705 DOI: 10.1371/journal.pone.0087858] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 12/31/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Insulin like growth factors (IGFs) and their binding proteins (IGFBPs) are secreted peptides that play major roles in regulating the normal development and maturation of mammary gland. While Igfbp7 has been shown to decrease breast tumor growth, its role in regulating the normal mammary gland development has not been studied. To this end, we generated Igfbp7-null mice and examined the development and maturation of mammary glands in the virgin, pregnant and lactating animals. RESULTS We report here that loss of Igfbp7 significantly retards mammary gland development in the virgin animals. More significantly, the pregnant Igfpb7-null glands contained fewer alveolar structures and that during lactation these glands exhibit the morphological changes that are associated with involution. The transcriptome profile of the Igfbp7-null glands on the lactation day 3 revealed a distinct involution-related gene signature compared to the lactating WT glands. Interestingly, we found that the lactating Igfbp7-null glands exhibit increased expression of Stat3 and enhanced activation of (phosphorylated) Stat3, combined with decreased expression of Stat5 suggesting that the absence of Igfbp7 accelerates the onset of involution. We also found that in absence of Igfpb7, the lactating glands contain increased Igfbp5 protein along with decreased expression of IGF-1 Receptor and Akt activation. Finally, we show that during the normal course of involution, Igfbp7 expression is significantly decreased in the mammary gland. CONCLUSION Our data suggest that loss of Igfbp7 induces precocious involution possibly through diminished cell survival signals. Our findings identify Igfbp7 as major regulator of involution in the mammary gland.
Collapse
|
36
|
Duitman J, Borensztajn KS, Pulskens WPC, Leemans JC, Florquin S, Spek CA. CCAAT-enhancer binding protein delta (C/EBPδ) attenuates tubular injury and tubulointerstitial fibrogenesis during chronic obstructive nephropathy. J Transl Med 2014; 94:89-97. [PMID: 24247561 DOI: 10.1038/labinvest.2013.127] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/02/2013] [Accepted: 10/08/2013] [Indexed: 12/30/2022] Open
Abstract
CCAAT-enhancer-binding protein delta (C/EBPδ) is a transcription factor mainly known for its role in inflammation and apoptosis/proliferation. Considering that these are key processes in renal fibrosis, we hypothesized that C/EBPδ would potentiate renal fibrosis. In line with this hypothesis, C/EBPδ has recently been suggested to regulate the fibrotic response during glomerulonephritis. Here we determined the importance of C/EBPδ in the development of renal tubulointerstitial fibrosis by subjecting 8- to 12-week-old C/EBPδ-deficient mice and age- and sex-matched wild-type controls to the unilateral ureteral obstruction model. Mice were killed at 1, 3, or 7 days post surgery, and renal tissues were obtained for RNA, protein, and immunohistochemical analysis. We show that C/EBPδ deficiency resulted in a more profound fibrotic response as evident from enhanced tubular injury, collagen deposition in the interstitial area, and higher expression of transforming growth factor-β. Moreover, we show that the increase in renal fibrosis in C/EBPδ-deficient mice does not depend on an altered proliferation/apoptosis balance or on a differential inflammatory response in the obstructed kidney. In conclusion, our study provides direct evidence that C/EBPδ is a novel mediator of renal fibrosis. Modulating C/EBPδ expression could consequently be a potential antifibrotic strategy in patients with chronic kidney disease.
Collapse
Affiliation(s)
- JanWillem Duitman
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Keren S Borensztajn
- Unité INSERM 700, Physiopathologie et Epidémiologie de l'Insuffisance Respiratoire, Faculté de Médecine Xavier Bichat, Paris, France
| | - Willem P C Pulskens
- Department of Physiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jaklien C Leemans
- Department of Pathology; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sandrine Florquin
- 1] Department of Pathology; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands [2] Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - C Arnold Spek
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Martinson HA, Lyons TR, Giles ED, Borges VF, Schedin P. Developmental windows of breast cancer risk provide opportunities for targeted chemoprevention. Exp Cell Res 2013; 319:1671-8. [PMID: 23664839 PMCID: PMC3980135 DOI: 10.1016/j.yexcr.2013.04.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 04/26/2013] [Accepted: 04/28/2013] [Indexed: 12/12/2022]
Abstract
The magnitude of the breast cancer problem implores researchers to aggressively investigate prevention strategies. However, several barriers currently reduce the feasibility of breast cancer prevention. These barriers include the inability to accurately predict future breast cancer diagnosis at the individual level, the need for improved understanding of when to implement interventions, uncertainty with respect to optimal duration of treatment, and negative side effects associated with currently approved chemoprevention therapies. None-the-less, the unique biology of the mammary gland, with its postnatal development and conditional terminal differentiation, may permit the resolution of many of these barriers. Specifically, lifecycle-specific windows of breast cancer risk have been identified that may be amenable to risk-reducing strategies. Here, we argue for prevention research focused on two of these lifecycle windows of risk: postpartum mammary gland involution and peri-menopause. We provide evidence that these windows are highly amenable to targeted, limited duration treatments. Such approaches could result in the prevention of postpartum and postmenopausal breast cancers, correspondingly.
Collapse
Affiliation(s)
- Holly A Martinson
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC1S, 8401K, 12801 East 17th Avenue, Aurora, CO 80045, USA; Program in Cancer Biology, University of Colorado Anschutz Medical Campus, MS8104, RC1S, 5117, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC1S, 8401K, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Erin D Giles
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus School of Medicine, RC1S, Room 7103, 12801 East 17th Avenue, Mail Stop 8106, Aurora, CO 80045, USA; Anschutz Health and Wellness Center, 12348 East Montview Boulevard, Campus Box C263, Aurora, CO 80045, USA
| | - Virginia F Borges
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC1S, 8401K, 12801 East 17th Avenue, Aurora, CO 80045, USA; University of Colorado Cancer Center, Building 500, Suite 6004C, 13001 East 17th Place, Aurora, CO 80045, USA; Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
| | - Pepper Schedin
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC1S, 8401K, 12801 East 17th Avenue, Aurora, CO 80045, USA; Program in Cancer Biology, University of Colorado Anschutz Medical Campus, MS8104, RC1S, 5117, 12801 East 17th Avenue, Aurora, CO 80045, USA; University of Colorado Cancer Center, Building 500, Suite 6004C, 13001 East 17th Place, Aurora, CO 80045, USA; Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA.
| |
Collapse
|
38
|
Site-specific CpG methylation in the CCAAT/enhancer binding protein delta (CEBPδ) CpG island in breast cancer is associated with metastatic relapse. Br J Cancer 2012; 107:732-8. [PMID: 22782348 PMCID: PMC3419957 DOI: 10.1038/bjc.2012.308] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background: The CCAAT/enhancer binding protein delta (CEBPδ) is a member of a highly conserved family of basic region leucine zipper transcription factors. It has properties consistent with a tumour suppressor; however, other data suggest that CEBPδ may be involved in the metastatic process. Methods: We analysed the expression of CEBPδ and the methylation status of the CpG island in human breast cancer cell lines, in 107 archival cases of primary breast cancer and in two series of metastatic breast cancers using qPCR and pyrosequencing. Results: Expression of CEBPδ is downregulated in primary breast cancer by site-specific methylation in the CEBPδ CpG island. Expression is also downregulated in 50% of cases during progression from primary carcinoma to metastatic lesions. The CEBPδ CpG island is methylated in 81% metastatic breast cancer lesions, while methylation in the CEBPδ CpG island in primary cancers is associated with increased risk of relapse and metastasis. Conclusion: CCAAT/enhancer binding protein delta CpG island methylation is associated with metastasis in breast cancer. Detection of methylated CEBPδ genomic DNA may have utility as an epigenetic biomarker of primary breast carcinomas at increased risk of relapse and metastasis.
Collapse
|
39
|
Borges VF, Schedin P. Could NSAIDs become a preventative therapy in pregnancy-associated breast cancer? BREAST CANCER MANAGEMENT 2012. [DOI: 10.2217/bmt.12.15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Pregnancy-associated breast cancer (PABC) is a unique type of young women’s breast cancer that includes two biologically distinct conditions: those diagnosed during pregnancy and those diagnosed postpartum. It is the dominant subset of postpartum PABC that is more consistently associated with higher breast cancer mortality. Preclinical work has identified the normal event of postpartum involution as a wound-healing milieu rich in immune cells. We have shown that the involution environment drives tumor growth, proliferation and metastasis. Moreover, we have demonstrated in animal models that this ‘involution effect’ can be abrogated with drug therapy, namely NSAIDs, which target normal involution pathways implicated in PABC tumor promotion. In this perspective, we review the contemporary understanding of PABC, our preclinical modeling and its implications and the unmet research needs required for future translation of these preclinical studies into rational and safe human clinical trials.
Collapse
Affiliation(s)
- Virginia F Borges
- University of Colorado Denver Anschutz Medical Campus, 12801 E. 17th Avenue, Room 8112, Aurora, CO 80045, USA
| | - Pepper Schedin
- AMC Cancer Research Center, 3401 Quebec Street, Suite 3200, Denver, CO 80207, USA
| |
Collapse
|
40
|
High fat diet alters lactation outcomes: possible involvement of inflammatory and serotonergic pathways. PLoS One 2012; 7:e32598. [PMID: 22403677 PMCID: PMC3293832 DOI: 10.1371/journal.pone.0032598] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 02/01/2012] [Indexed: 11/23/2022] Open
Abstract
Delay in the onset of lactogenesis has been shown to occur in women who are obese, however the mechanism altered within the mammary gland causing the delay remains unknown. Consumption of high fat diets (HFD) has been previously determined to result decreased litters and litter numbers in rodent models due to a decrease in fertility. We examined the effects of feeding a HFD (60% kcal from fat) diet versus a low-fat diet (LFD; 10% kcal from fat) to female Wistar rats on lactation outcomes. Feeding of HFD diet resulted in increased pup weights compared to pups from LFD fed animals for 4 d post-partum. Lactation was delayed in mothers on HFD but they began to produce copious milk volumes beginning 2 d post-partum, and milk yield was similar to LFD by day 3. Mammary glands collected from lactating animals on HFD diet, displayed a disrupted morphologies, with very few and small alveoli. Consistently, there was a significant decrease in the mRNA expression of milk protein genes, glucose transporter 1 (GLUT1) and keratin 5 (K5), a luminobasal cell marker in the mammary glands of HFD lactating animals. Expression of tryptophan hydroxylase 1 (TPH1), the rate-limiting enzyme in serotonin (5-HT) biosynthesis, and the 5-HT7 receptor (HTR7), which regulates mammary gland involution, were significantly increased in mammary glands of HFD animals. Additionally, we saw elevation of the inflammatory markers interleukin-6 (IL-6) and tumor necrosis factor-α (TNF- α). These results indicate that consumption of HFD impairs mammary parenchymal tissue and impedes its ability to synthesize and secrete milk, possibly through an increase in 5-HT production within the mammary gland leading to an inflammatory process.
Collapse
|
41
|
Güllü G, Karabulut S, Akkiprik M. Functional roles and clinical values of insulin-like growth factor-binding protein-5 in different types of cancers. CHINESE JOURNAL OF CANCER 2012; 31:266-80. [PMID: 22313597 PMCID: PMC3777492 DOI: 10.5732/cjc.011.10405] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insulin-like growth factor-binding proteins (IGFBPs) are critical regulators of the mitogenic activity of insulin-like growth factors (IGFs). IGFBP5, one of these IGFBPs, has special structural features, including a nuclear transport domain, heparin-binding motif, and IGF/extracellular matrix/acid-labile subunit-binding sites. Furthermore, IGFBP5 has several functional effects on carcinogenesis and even normal cell processes, such as cell growth, death, motility, and tissue remodeling. These biological effects are sometimes related with IGF (IGF-dependent effects) and sometimes not (IGF-independent effects). The functional role of IGFBP5 is most likely determined in a cell-type and tissue-type specific manner but also depends on cell context, especially in terms of the diversity of interacting proteins and the potential for nuclear localization. Clinical findings show that IGFBP5 has the potential to be a useful clinical biomarker for predicting response to therapy and clinical outcome of cancer patients. In this review, we summarize the functional diversity and clinical importance of IGFBP5 in different types of cancers.
Collapse
Affiliation(s)
- Gökçe Güllü
- Department of Medical Biology, School of Medicine, DMarmara University, Istanbul 34468, Turkey
| | | | | |
Collapse
|
42
|
Barker HE, Chang J, Cox TR, Lang G, Bird D, Nicolau M, Evans HR, Gartland A, Erler JT. LOXL2-mediated matrix remodeling in metastasis and mammary gland involution. Cancer Res 2011; 71:1561-72. [PMID: 21233336 PMCID: PMC3842018 DOI: 10.1158/0008-5472.can-10-2868] [Citation(s) in RCA: 216] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
More than 90% of cancer patient mortality is attributed to metastasis. In this study, we investigated a role for the lysyl oxidase-related enzyme lysyl oxidase-like 2 (LOXL2) in breast cancer metastasis, in both patient samples and in vivo models. Analysis of a published microarray data set revealed that LOXL2 expression is correlated with metastasis and decreased survival in patients with aggressive breast cancer. In immunocompetent or immunocompromised orthotopic and transgenic breast cancer models we showed that genetic, chemical or antibody-mediated inhibition of LOXL2 resulted in decreased metastasis. Mechanistic investigations revealed that LOXL2 promotes invasion by regulating the expression and activity of the extracellular proteins tissue inhibitor of metalloproteinase-1 (TIMP1) and matrix metalloproteinase-9 (MMP9). We found that LOXL2, TIMP1, and MMP9 are coexpressed during mammary gland involution, suggesting they function together in glandular remodeling after weaning. Finally, we found that LOXL2 is highly expressed in the basal/myoepithelial mammary cell lineage, like many other genes that are upregulated in basal-like breast cancers. Our findings highlight the importance of LOXL2 in breast cancer progression and support the development of anti-LOXL2 therapeutics for the treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Holly E. Barker
- Section of Cell and Molecular Biology, The Institute of Cancer Research, London SW3 6JB, UK
| | - Joan Chang
- Section of Cell and Molecular Biology, The Institute of Cancer Research, London SW3 6JB, UK
| | - Thomas R Cox
- Section of Cell and Molecular Biology, The Institute of Cancer Research, London SW3 6JB, UK
- Cancer Research UK Tumour Cell Signalling Unit, The Institute of Cancer Research, London SW3 6JB, UK
| | - Georgina Lang
- Section of Cell and Molecular Biology, The Institute of Cancer Research, London SW3 6JB, UK
- Cancer Research UK Tumour Cell Signalling Unit, The Institute of Cancer Research, London SW3 6JB, UK
| | - Demelza Bird
- Section of Cell and Molecular Biology, The Institute of Cancer Research, London SW3 6JB, UK
- Cancer Research UK Tumour Cell Signalling Unit, The Institute of Cancer Research, London SW3 6JB, UK
| | - Monica Nicolau
- Department of Mathematics, Stanford University, Stanford CA 94305, USA
| | - Holly R. Evans
- The Mellanby Centre For Bone Research, Department of Human Metabolism, The University of Sheffield, Sheffield S10 2RX, UK
| | - Alison Gartland
- The Mellanby Centre For Bone Research, Department of Human Metabolism, The University of Sheffield, Sheffield S10 2RX, UK
| | - Janine T. Erler
- Section of Cell and Molecular Biology, The Institute of Cancer Research, London SW3 6JB, UK
- Cancer Research UK Tumour Cell Signalling Unit, The Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
43
|
Veiseh M, Turley EA. Hyaluronan metabolism in remodeling extracellular matrix: probes for imaging and therapy of breast cancer. Integr Biol (Camb) 2011; 3:304-15. [PMID: 21264398 DOI: 10.1039/c0ib00096e] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Clinical and experimental evidence increasingly support the concept of cancer as a disease that emulates a component of wound healing, in particular abnormal stromal extracellular matrix remodeling. Here we review the biology and function of one remodeling process, hyaluronan (HA) metabolism, which is essential for wound resolution but closely linked to breast cancer (BCA) progression. Components of the HA metabolic cycle (HAS2, SPAM1 and HA receptors CD44, RHAMM/HMMR and TLR2) are discussed in terms of their known functions in wound healing and in breast cancer progression. Finally, we discuss recent advances in the use of HA-based platforms for developing nanoprobes to image areas of active HA metabolism and for therapeutics in breast cancer.
Collapse
Affiliation(s)
- M Veiseh
- Life Sciences Division, Lawrence Berkeley National Laboratories, Berkeley, CA, USA.
| | | |
Collapse
|
44
|
Deregulation of HIF1-alpha and hypoxia-regulated pathways in hepatocellular carcinoma and corresponding non-malignant liver tissue--influence of a modulated host stroma on the prognosis of HCC. Langenbecks Arch Surg 2010; 395:395-405. [PMID: 20165955 DOI: 10.1007/s00423-009-0590-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2009] [Accepted: 12/22/2009] [Indexed: 12/20/2022]
Abstract
PURPOSE The aim of this study was to elucidate the role of HIF1A expression in hepatocellular carcinoma (HCC) and the corresponding non-malignant liver tissue and to correlate it with the clinical outcome of HCC patients after curative liver resection. METHODS HIF1A expression was determined by quantitative RT-PCR in HCC and corresponding non-malignant liver tissue of 53 patients surgically treated for HCC. High-density gene expression analysis and pathway analysis was performed on a selected subset of patients with high and low HIF1A expression in the non-malignant liver tissue. RESULTS HIF1A over-expression in the apparently non-malignant liver tissue was a predictor of tumor recurrence and survival. The estimated 1-year and 5-year disease-free survival was significantly better in patients with low HIF1A expression in the non-malignant liver tissue when compared to those patients with high HIF1 expression (88.9% vs. 67.9% and 61.0% vs. 22.6%, respectively, p = 0.008). Based on molecular pathway analysis utilizing high-density gene-expression profiling, HIF1A related molecular networks were identified that contained genes involved in cell migration, cell homing, and cell-cell interaction. CONCLUSION Our study identified a potential novel mechanism contributing to prognosis of HCC. The deregulation of HIF1A and its related pathways in the apparently non-malignant liver tissue provides for a modulated environment that potentially enhances or allows for HCC recurrence after curative resection.
Collapse
|