1
|
Singh P, Ali SA. Mature white adipocyte plasticity during mammary gland remodelling and cancer. CELL INSIGHT 2023; 2:100123. [PMID: 37771567 PMCID: PMC10522874 DOI: 10.1016/j.cellin.2023.100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/30/2023]
Abstract
Mammary gland growth and differentiation predominantly rely on stromal-epithelial cellular communication. Specifically, mammary adipocytes play a crucial role in ductal morphogenesis, as well as in the proliferation and differentiation of mammary epithelial cells. The process of lactation entails a reduction in the levels of white adipose tissue associated with the MG, allowing for the expansion of milk-producing epithelial cells. Subsequently, during involution and the regression of the milk-producing unit, adipocyte layers resurface, occupying the vacated space. This dynamic phenomenon underscores the remarkable plasticity and expansion of adipose tissue. Traditionally considered terminally differentiated, adipocytes have recently been found to exhibit plasticity in certain contexts. Unraveling the significance of this cell type within the MG could pave the way for novel approaches to reduce the risk of breast cancer and enhance lactation performance. Moreover, a comprehensive understanding of adipocyte trans- and de-differentiation processes holds promise for the development of innovative therapeutic interventions targeting cancer, fibrosis, obesity, type 2 diabetes, and other related diseases. Additionally, adipocytes may find utility in the realm of regenerative medicine. This review article provides a comprehensive examination of recent advancements in our understanding of MG remodelling, with a specific focus on the tissue-specific functions of adipocytes and their role in the development of cancer. By synthesizing current knowledge in this field, it aims to consolidate our understanding of adipocyte biology within the context of mammary gland biology, thereby fostering further research and discovery in this vital area.
Collapse
Affiliation(s)
- Parul Singh
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division Proteomics of Stem Cells and Cancer, German Cancer Research Center, 69120, Heidelberg, Germany
| |
Collapse
|
2
|
Horst EA, Kvidera SK, Baumgard LH. Invited review: The influence of immune activation on transition cow health and performance-A critical evaluation of traditional dogmas. J Dairy Sci 2021; 104:8380-8410. [PMID: 34053763 DOI: 10.3168/jds.2021-20330] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022]
Abstract
The progression from gestation into lactation represents the transition period, and it is accompanied by marked physiological, metabolic, and inflammatory adjustments. The entire lactation and a cow's opportunity to have an additional lactation are heavily dependent on how successfully she adapts during the periparturient period. Additionally, a disproportionate amount of health care and culling occurs early following parturition. Thus, lactation maladaptation has been a heavily researched area of dairy science for more than 50 yr. It was traditionally thought that excessive adipose tissue mobilization in large part dictated transition period success. Further, the magnitude of hypocalcemia has also been assumed to partly control whether a cow effectively navigates the first few months of lactation. The canon became that adipose tissue released nonesterified fatty acids (NEFA) and the resulting hepatic-derived ketones coupled with hypocalcemia lead to immune suppression, which is responsible for transition disorders (e.g., mastitis, metritis, retained placenta, poor fertility). In other words, the dogma evolved that these metabolites and hypocalcemia were causal to transition cow problems and that large efforts should be enlisted to prevent increased NEFA, hyperketonemia, and subclinical hypocalcemia. However, despite intensive academic and industry focus, the periparturient period remains a large hurdle to animal welfare, farm profitability, and dairy sustainability. Thus, it stands to reason that there are alternative explanations to periparturient failures. Recently, it has become firmly established that immune activation and the ipso facto inflammatory response are a normal component of transition cow biology. The origin of immune activation likely stems from the mammary gland, tissue trauma during parturition, and the gastrointestinal tract. If inflammation becomes pathological, it reduces feed intake and causes hypocalcemia. Our tenet is that immune system utilization of glucose and its induction of hypophagia are responsible for the extensive increase in NEFA and ketones, and this explains why they (and the severity of hypocalcemia) are correlated with poor health, production, and reproduction outcomes. In this review, we argue that changes in circulating NEFA, ketones, and calcium are simply reflective of either (1) normal homeorhetic adjustments that healthy, high-producing cows use to prioritize milk synthesis or (2) the consequence of immune activation and its sequelae.
Collapse
Affiliation(s)
- E A Horst
- Department of Animal Science, Iowa State University, Ames 50011
| | - S K Kvidera
- Department of Animal Science, Iowa State University, Ames 50011
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames 50011.
| |
Collapse
|
3
|
Verma AK, Ali SA, Singh P, Kumar S, Mohanty AK. Transcriptional Repression of MFG-E8 Causes Disturbance in the Homeostasis of Cell Cycle Through DOCK/ZP4/STAT Signaling in Buffalo Mammary Epithelial Cells. Front Cell Dev Biol 2021; 9:568660. [PMID: 33869165 PMCID: PMC8047144 DOI: 10.3389/fcell.2021.568660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
The mammary gland is a unique apocrine gland made up of a branching network of ducts that end in alveoli. It is an ideal system to study the molecular mechanisms associated with cell proliferation, differentiation, and oncogenesis. MFG-E8, also known as Lactadherin, is a vital glycoprotein related to the milk fat globule membrane and initially identified to get secreted in bovine milk. Our previous report suggests that a high level of MFG-E8 is indicative of high milk yield in dairy animals. Here, we showed that MFG-E8 controls the cell growth and morphology of epithelial cells through a network of regulatory transcription factors. To understand the comprehensive action, we downregulated its expression in MECs by MFG-E8 specific shRNA. We generated a knockdown proteome profile of differentially expressed proteins through a quantitative iTRAQ experiment on a high-resolution mass spectrometer (Q-TOF). The downregulation of MFG-E8 resulted in reduced phagocytosis and cell migration ability, whereas it also leads to more lifespan to knockdown vis-a-vis healthy cells, which is confirmed through BrdU, MTT, and Caspase 3/7. The bioinformatics analysis revealed that MFG-E8 knockdown perturbs a large number of intracellular signaling, eventually leading to cessation in cell growth. Based on the directed network analysis, we found that MFG-E8 is activated by CX3CL1, TP63, and CSF2 and leads to the activation of SOCS3 and CCL2 for the regulation of cell proliferation. We further proved that the depletion of MFG-E8 resulted in activated cytoskeletal remodeling by MFG-E8 knockdown, which results in the activation of three independent pathways ZP4/JAK-STAT5, DOCK1/STAT3, and PIP3/AKT/mTOR. Overall, this study suggests that MFG-E8 expression in mammary epithelial cells is an indication of intracellular deterioration in cell health. To date, to the best of our knowledge, this is the first study that explores the downstream targets of MFG-E8 involved in the regulation of mammary epithelial cell health.
Collapse
|
4
|
Elder AM, Stoller AR, Black SA, Lyons TR. Macphatics and PoEMs in Postpartum Mammary Development and Tumor Progression. J Mammary Gland Biol Neoplasia 2020; 25:103-113. [PMID: 32535810 PMCID: PMC7395889 DOI: 10.1007/s10911-020-09451-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
Postpartum mammary gland involution is a mammalian tissue remodeling event that occurs after pregnancy and lactation to return the gland to the pre-pregnant state. This event is characterized by apoptosis and lysosomal-mediated cell death of the majority of the lactational mammary epithelium, followed by remodeling of the extracellular matrix, influx of immune cell populations (in particular, T helper cells, monocytes, and macrophages), and neo-lymphangiogenesis. This postpartum environment has been shown to be promotional for tumor growth and metastases and may partially account for why women diagnosed with breast cancer during the postpartum period or within 5 years of last childbirth have an increased risk of developing metastases when compared to their nulliparous counterparts. The lymphatics and macrophages present during mammary gland involution have been implicated in promoting the observed growth and metastasis. Of importance are the macrophages, which are of the "M2" phenotype and are known to create a pro-tumor microenvironment. In this report, we describe a subset of postpartum macrophages that express lymphatic proteins (PoEMs) and directly interact with lymphatic vessels to form chimeric vessels or "macphatics". Additionally, these PoEMs are very similar to tumor-associated macrophages that also express lymphatic proteins and are present at the sites of lymphatic vessels where tumors escape the tissue and enter the lymphatic vasculature. Further characterizing these PoEMs may offer insight in preventing lymphatic metastasis of breast cancer, as well as provide information for how developmental programming of lymphatic endothelial cells and macrophages can contribute to different disease progression.
Collapse
Affiliation(s)
- Alan M Elder
- Young Women's Breast Cancer Translational Program, Division of Medical Oncology, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, Aurora, CO, 80045, USA
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA
- Graduate Program in Cancer Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexander R Stoller
- Young Women's Breast Cancer Translational Program, Division of Medical Oncology, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, Aurora, CO, 80045, USA
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA
| | - Sarah A Black
- University of Colorado School of Medicine, Aurora, CO, USA
| | - Traci R Lyons
- Young Women's Breast Cancer Translational Program, Division of Medical Oncology, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, Aurora, CO, 80045, USA.
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA.
- Graduate Program in Cancer Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
5
|
Hop GE, de Prado-Taranilla AI, Isaka N, Ocak M, Bertet J, Supré K, Velthuis A, Schukken YH, Deflandre A. Efficacy of cabergoline in a double-blind randomized clinical trial on milk leakage reduction at drying-off and new intramammary infections across the dry period and postcalving. J Dairy Sci 2019; 102:11670-11680. [PMID: 31521356 DOI: 10.3168/jds.2019-16281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/14/2019] [Indexed: 12/25/2022]
Abstract
The abrupt cessation of milking at dry-off may induce milk leakage, which may increase the risk of new intramammary infections (IMI). This study assessed the efficacy of 1 i.m. injection of 5.6 mg of cabergoline (Velactis, Ceva Santé Animale, Libourne, France) at drying-off on milk leakage after dry-off and new IMI across the dry period and postcalving compared with a placebo (negative control) and an intramammary antibiotic treatment (positive control) under field conditions. The study was a double-blind, randomized, 3-arm, multicenter, clinical trial performed under Good Clinical Practice conditions. Data from 900 dairy cows of various breeds from 63 farms in France, Germany, and Hungary were analyzed. Only quarters with no bacterial growth at drying-off and a cow somatic cell count ≤200,000 cells/mL were included. Quarters infected with major or minor pathogens or cows with high somatic cell count at time of inclusion were excluded. Cows that qualified for the study were visited 7 times in total before and after drying-off and after calving. Presence (yes/no) of milk leakage was recorded on the day after dry-off. A new infected quarter (new IMI) was defined as one with a major pathogen present in any one of the 2 postcalving samples. Two mixed logistic regression models were fitted to the data to evaluate the efficacy of cabergoline in the reduction of milk leakage and new IMI. One i.m. injection of cabergoline at drying-off significantly reduced the incidence of milk leakage the day after dry-off compared with both placebo and antibiotic treatment. Cabergoline-treated cows significantly reduced the risk of new IMI by major pathogens across the dry period and postcalving by 21% when compared with placebo cows (20.5 vs. 26.0%, respectively). However, when milk leakage was added to the model, the significance of cabergoline was reduced. We interpreted this to show that milk leakage is an intervening variable between treatment with cabergoline and lower risk of new IMI. The antibiotic treatment significantly decreased the odds of new IMI compared with both cabergoline and placebo. However, because several countries are currently disallowing the preventive use of antibiotics at dry-off in noninfected quarters, the dry-off facilitator cabergoline may therefore be of particular value to reduce the risk of new IMI across the dry period.
Collapse
Affiliation(s)
- G E Hop
- GD Animal Health, PO Box 9, 7400 AA Deventer, the Netherlands
| | | | - N Isaka
- CEVA Santé Animale, Avenue de la Ballastière, 33500 Libourne, France
| | - M Ocak
- MD Research, An der Isar, 82049 Pullach im Isartal, Germany
| | - J Bertet
- CEVA Santé Animale, Avenue de la Ballastière, 33500 Libourne, France
| | - K Supré
- Milk Control Centre Flanders, Hagenbroeksesteenweg, B-2500 Lier, Belgium
| | - A Velthuis
- GD Animal Health, PO Box 9, 7400 AA Deventer, the Netherlands
| | - Y H Schukken
- GD Animal Health, PO Box 9, 7400 AA Deventer, the Netherlands
| | - A Deflandre
- CEVA Santé Animale, Avenue de la Ballastière, 33500 Libourne, France
| |
Collapse
|
6
|
Role of the Microenvironment in Regulating Normal and Cancer Stem Cell Activity: Implications for Breast Cancer Progression and Therapy Response. Cancers (Basel) 2019; 11:cancers11091240. [PMID: 31450577 PMCID: PMC6770706 DOI: 10.3390/cancers11091240] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/16/2019] [Accepted: 08/19/2019] [Indexed: 12/12/2022] Open
Abstract
The epithelial cells in an adult woman’s breast tissue are continuously replaced throughout their reproductive life during pregnancy and estrus cycles. Such extensive epithelial cell turnover is governed by the primitive mammary stem cells (MaSCs) that proliferate and differentiate into bipotential and lineage-restricted progenitors that ultimately generate the mature breast epithelial cells. These cellular processes are orchestrated by tightly-regulated paracrine signals and crosstalk between breast epithelial cells and their tissue microenvironment. However, current evidence suggests that alterations to the communication between MaSCs, epithelial progenitors and their microenvironment plays an important role in breast carcinogenesis. In this article, we review the current knowledge regarding the role of the breast tissue microenvironment in regulating the special functions of normal and cancer stem cells. Understanding the crosstalk between MaSCs and their microenvironment will provide new insights into how an altered breast tissue microenvironment could contribute to breast cancer development, progression and therapy response and the implications of this for the development of novel therapeutic strategies to target cancer stem cells.
Collapse
|
7
|
Zhao X, Ponchon B, Lanctôt S, Lacasse P. Invited review: Accelerating mammary gland involution after drying-off in dairy cattle. J Dairy Sci 2019; 102:6701-6717. [PMID: 31202662 DOI: 10.3168/jds.2019-16377] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/22/2019] [Indexed: 01/20/2023]
Abstract
Bovine mammary gland involution, as a part of the reproductive cycle in dairy cows, is a very important remodeling transformation of the mammary gland for the subsequent lactation. There is considerable incentive to accelerate mammary gland involution to improve udder health, shorten the dry period, and simplify the management process by reducing dietary changes. The complex process of mammary involution is characterized by morphological changes in the epithelial cells and mammary tissue, changes in the composition of mammary secretions, and changes in the integrity of tight junctions. Involution is facilitated by elements of the immune system and several types of proteases and is coordinated by various types of hormones. This review first describes the involution process and then argues for the need to accelerate it. Last, this review focuses on various intervention methods for accelerating involution. Our aim is to provide a comprehensive overview of bovine mammary gland involution as well as potential techniques and new opinions for dry cow management.
Collapse
Affiliation(s)
- X Zhao
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada H9X 3V9.
| | - B Ponchon
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada H9X 3V9
| | - S Lanctôt
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada J1M 0C8
| | - P Lacasse
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada J1M 0C8
| |
Collapse
|
8
|
Santucci-Pereira J, Zeleniuch-Jacquotte A, Afanasyeva Y, Zhong H, Slifker M, Peri S, Ross EA, López de Cicco R, Zhai Y, Nguyen T, Sheriff F, Russo IH, Su Y, Arslan AA, Bordas P, Lenner P, Åhman J, Landström Eriksson AS, Johansson R, Hallmans G, Toniolo P, Russo J. Genomic signature of parity in the breast of premenopausal women. Breast Cancer Res 2019; 21:46. [PMID: 30922380 PMCID: PMC6438043 DOI: 10.1186/s13058-019-1128-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/14/2019] [Indexed: 12/17/2022] Open
Abstract
Background Full-term pregnancy (FTP) at an early age confers long-term protection against breast cancer. Previously, we reported that a FTP imprints a specific gene expression profile in the breast of postmenopausal women. Herein, we evaluated gene expression changes induced by parity in the breast of premenopausal women. Methods Gene expression profiling of normal breast tissue from 30 nulliparous (NP) and 79 parous (P) premenopausal volunteers was performed using Affymetrix microarrays. In addition to a discovery/validation analysis, we conducted an analysis of gene expression differences in P vs. NP women as a function of time since last FTP. Finally, a laser capture microdissection substudy was performed to compare the gene expression profile in the whole breast biopsy with that in the epithelial and stromal tissues. Results Discovery/validation analysis identified 43 differentially expressed genes in P vs. NP breast. Analysis of expression as a function of time since FTP revealed 286 differentially expressed genes (238 up- and 48 downregulated) comparing all P vs. all NP, and/or P women whose last FTP was less than 5 years before biopsy vs. all NP women. The upregulated genes showed three expression patterns: (1) transient: genes upregulated after FTP but whose expression levels returned to NP levels. These genes were mainly related to immune response, specifically activation of T cells. (2) Long-term changing: genes upregulated following FTP, whose expression levels decreased with increasing time since FTP but did not return to NP levels. These were related to immune response and development. (3) Long-term constant: genes that remained upregulated in parous compared to nulliparous breast, independently of time since FTP. These were mainly involved in development/cell differentiation processes, and also chromatin remodeling. Lastly, we found that the gene expression in whole tissue was a weighted average of the expression in epithelial and stromal tissues. Conclusions Genes transiently activated by FTP may have a role in protecting the mammary gland against neoplastically transformed cells through activation of T cells. Furthermore, chromatin remodeling and cell differentiation, represented by the genes that are maintained upregulated long after the FTP, may be responsible for the lasting preventive effect against breast cancer. Electronic supplementary material The online version of this article (10.1186/s13058-019-1128-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Santucci-Pereira
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA.
| | - Anne Zeleniuch-Jacquotte
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA.,New York University Perlmutter Cancer Center, New York, NY, 10016, USA
| | - Yelena Afanasyeva
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA
| | - Hua Zhong
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA
| | - Michael Slifker
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center - Temple University Health System, Philadelphia, PA, 19111, USA
| | - Suraj Peri
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center - Temple University Health System, Philadelphia, PA, 19111, USA
| | - Eric A Ross
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center - Temple University Health System, Philadelphia, PA, 19111, USA
| | - Ricardo López de Cicco
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Yubo Zhai
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Theresa Nguyen
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Fathima Sheriff
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Irma H Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Yanrong Su
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Alan A Arslan
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA.,Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY, 10016, USA
| | - Pal Bordas
- Sunderby Hospital, Luleå and the Norrbotten Mammography Screening Program, Luleå, Sweden.,Departments of Radiation Sciences and Oncology, Umeå University, Umeå, Sweden
| | - Per Lenner
- Departments of Radiation Sciences and Oncology, Umeå University, Umeå, Sweden
| | - Janet Åhman
- Sunderby Hospital, Luleå and the Norrbotten Mammography Screening Program, Luleå, Sweden
| | | | | | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Paolo Toniolo
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY, 10016, USA
| | - Jose Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| |
Collapse
|
9
|
Mammary Stem Cells in Domestic Animals: The Role of ROS. Antioxidants (Basel) 2018; 8:antiox8010006. [PMID: 30587765 PMCID: PMC6356801 DOI: 10.3390/antiox8010006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/12/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) are produced as a natural byproduct of the normal metabolism of oxygen and play significant roles in cell signaling and homeostasis. Although ROS have been involved in pathological processes as diverse as cancer, cardiovascular disease, and aging, they may to exert an effect even in a physiological context. In the central nervous system, stem cells and hematopoietic stem cells are early progenitors that contain lower levels of ROS than their more mature progeny. These different concentrations have been reported to be crucial for maintaining stem cell function. Mammary gland remodeling has been proposed to be organized through the activation and regulation of cells with stemness, either considered real stem cells or primitive precursors. Given the state of oxidative stress in the mammary gland tissue induced by high milk production, in particular in highly productive dairy cows; several studies have focused on the relationship between adult mammary stem cells and the oxidative state of the gland. The oxidative state of the mammary gland appears to be involved in the initial development and metastasis of breast cancer through interference with mammary cancerous stem cells. This review summarizes some links between the mammary stem and oxidative state of the gland.
Collapse
|
10
|
Stromal cells in breast cancer as a potential therapeutic target. Oncotarget 2018; 9:23761-23779. [PMID: 29805773 PMCID: PMC5955086 DOI: 10.18632/oncotarget.25245] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
Breast cancer in the United States is the second most commonly diagnosed cancer in women. About 1 in 8 women will develop invasive breast cancer over the course of her lifetime and breast cancer remains the second leading cause of cancer-related death. In pursuit of novel therapeutic strategies, researchers have examined the tumor microenvironment as a potential anti-cancer target. In addition to neoplastic cells, the tumor microenvironment is composed of several critical normal cell types, including fibroblasts, vascular and lymph endothelial cells, osteoclasts, adipocytes, and immune cells. These cells have important roles in healthy tissue stasis, which frequently are altered in tumors. Indeed, tumor-associated stromal cells often contribute to tumorigenesis, tumor progression, and metastasis. Consequently, these host cells may serve as a possible target in anti-tumor and anti-metastatic therapeutic strategies. Targeting the tumor associated host cells offers the benefit that such cells do not mutate and develop resistance in response to treatment, a major cause of failure in cancer therapeutics targeting neoplastic cells. This review discusses the role of host cells in the tumor microenvironment during tumorigenesis, progression, and metastasis, and provides an overview of recent developments in targeting these cell populations to enhance cancer therapy efficacy.
Collapse
|
11
|
Ismail OZ, Sriranganathan S, Zhang X, Bonventre JV, Zervos AS, Gunaratnam L. Tctex-1, a novel interaction partner of Kidney Injury Molecule-1, is required for efferocytosis. J Cell Physiol 2018; 233:6877-6895. [PMID: 29693725 DOI: 10.1002/jcp.26578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 03/01/2018] [Indexed: 02/04/2023]
Abstract
Kidney injury molecule-1 (KIM-1) is a phosphatidylserine receptor that is specifically upregulated on proximal tubular epithelial cells (PTECs) during acute kidney injury and mitigates tissue damage by mediating efferocytosis (the phagocytic clearance of apoptotic cells). The signaling molecules that regulate efferocytosis in TECs are not well understood. Using a yeast two-hybrid screen, we identified the dynein light chain protein, Tctex-1, as a novel KIM-1-interacting protein. Immunoprecipitation and confocal imaging studies suggested that Tctex-1 associates with KIM-1 in cells at baseline, but, dissociates from KIM-1 within 90 min of initiation of efferocytosis. Interfering with actin or microtubule polymerization interestingly prevented the dissociation of KIM-1 from Tctex-1. Moreover, the subcellular localization of Tctex-1 changed from being microtubule-associated to mainly cytosolic upon expression of KIM-1. Short hairpin RNA-mediated silencing of endogenous Tctex-1 in cells significantly inhibited efferocytosis to levels comparable to that of knock down of KIM-1 in the same cells. Importantly, Tctex-1 was not involved in the delivery of KIM-1 to the cell-surface. On the other hand, KIM-1 expression significantly inhibited the phosphorylation of Tctex-1 at threonine 94 (T94), a post-translational modification which is known to disrupt the binding of Tctex-1 to dynein on microtubules. In keeping with this, we found that KIM-1 bound less efficiently to the phosphomimic (T94E) mutant of Tctex-1 compared to wild type Tctex-1. Surprisingly, expression of Tctex-1 T94E did not influence KIM-1-mediated efferocytosis. Our studies uncover a previously unknown role for Tctex-1 in KIM-1-dependent efferocytosis in epithelial cells.
Collapse
Affiliation(s)
- Ola Z Ismail
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Lawson Health Research Institute, London, Ontario, Canada
| | - Saranga Sriranganathan
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Lawson Health Research Institute, London, Ontario, Canada
| | - Xizhong Zhang
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Lawson Health Research Institute, London, Ontario, Canada
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Antonis S Zervos
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, Florida
| | - Lakshman Gunaratnam
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Lawson Health Research Institute, London, Ontario, Canada.,Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, London, Western University, Ontario, Canada
| |
Collapse
|
12
|
ElShamy WM. The protective effect of longer duration of breastfeeding against pregnancy-associated triple negative breast cancer. Oncotarget 2018; 7:53941-53950. [PMID: 27248476 PMCID: PMC5288234 DOI: 10.18632/oncotarget.9690] [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: 04/09/2016] [Accepted: 05/23/2016] [Indexed: 12/24/2022] Open
Abstract
Parity associated breast cancer (PABC) often diagnosed within the 2-5 years after a full term pregnancy. PABC is usually present with more advanced, poorly differentiated, high-grade cancers that show shorter time to progression and often of the triple negative breast cancer (TNBC) subtype. Data from around the world show that pregnancy-associated TNBC is independently associated with poor survival, underscoring the impact of the pregnant breast microenvironment on the biology and consequently the prognosis of these tumors. Although it is not yet clear, a link between pregnancy-associated TNBCs and lack or shorter duration of breastfeeding (not pregnancy per se) has been proposed. Here, we present epidemiological and experimental evidence for the protective effect of longer duration of lactation against pregnancy-associated TNBCs, and propose a putative molecular mechanism for this protective effect and its effect in eliminating any potential TNBC precursors from the breast by the end of the natural breast involution.
Collapse
Affiliation(s)
- Wael M ElShamy
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
13
|
Ponchon B, Lacasse P, Ollier S, Zhao X. Effects of photoperiod modulation and melatonin feeding around drying-off on bovine mammary gland involution. J Dairy Sci 2017; 100:8496-8506. [PMID: 28755938 DOI: 10.3168/jds.2016-12272] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 05/29/2017] [Indexed: 01/01/2023]
Abstract
The risk for a dairy cow to acquire new intramammary infections is high during the transition from lactation to the dry period, because of udder engorgement and altered immune functions. Once the gland is fully involuted, it becomes much more resistant to intramammary infections. Therefore, strategies to depress milk yield before drying-off and accelerate the involution process after drying-off could be beneficial for udder health. The objective of this study was to assess the effect of photoperiod manipulation and melatonin feeding from 14 d before to 14 d after drying-off on the speed of the involution process. Thirty Holstein cows in late lactation were randomly allocated to one of the following treatments: (1) a long-day photoperiod (16 h of light: 8 h of darkness), (2) a short-day photoperiod (8 h of light: 16 h of darkness), and (3) a long-day photoperiod supplemented by melatonin feeding (4 mg/kg of body weight). Milk and blood samples were collected on d -26, -19, -12, -5, -1, 1, 3, 5, 7, 10, and 14 relative to the last milking to determine concentrations of mammary gland involution markers and serum prolactin. Additional blood samples were taken around milking on d -15, before the start of the treatments, and on d -1, before drying-off, to evaluate the treatment effects on milking-induced prolactin release. The short-day photoperiod slightly decreased milk production and basal prolactin secretion during the dry period. The milking-induced prolactin surge was smaller on d -1 than on d -15 regardless of the treatments. Lactoferrin concentration, somatic cell count, and BSA concentration as well as matrix metalloproteinase-2 and -9 activities increased in mammary secretions during the first 2 wk of the dry period, whereas milk citrate concentration and the citrate:lactoferrin molar ratio decreased. The rates of change of these parameters were not significantly affected by the treatments. The long-day photoperiod supplemented by melatonin feeding did not affect milk production, prolactin secretion, or mammary gland involution. Under the conditions in this study, photoperiod modulation and melatonin feeding did not appear to affect the rate of mammary gland involution.
Collapse
Affiliation(s)
- B Ponchon
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada H9X 3V9
| | - P Lacasse
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Quebec, Canada J1M 0C8
| | - S Ollier
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Quebec, Canada J1M 0C8
| | - X Zhao
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada H9X 3V9.
| |
Collapse
|
14
|
Farmer C, Amezcua M, Bruckmaier R, Wellnitz O, Friendship R. Does duration of teat use in first parity affect milk yield and mammary gene expression in second parity? J Anim Sci 2017; 95:681-687. [PMID: 28380616 DOI: 10.2527/jas.2016.1119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
It was recently shown that a teat that is not used in the first lactation will have a reduced development and milk yield in the second lactation. In the current study, the impact of imposing a suckling period of 2, 7, or 21 d during the first lactation on piglet performance, milk composition, endocrine status, and mammary gene expression of sows in their second lactation was studied. Pregnant Yorkshire gilts were divided into 3 groups according to lactation length: 1) 2-d lactation (2D; = 20), 2) 7-d lactation (7D; = 20), and 3) 21-d lactation (21D; = 21). After weaning, sows were bred and kept for a second parity. In both lactations, litters were standardized to 12 piglets with 12 functional teats and surplus teats were sealed. In the second lactation, piglets were weighed on d 2, 7, 14, 21 (weaning), 31, and 56 postpartum, and sow feed intake was recorded. On d 110 of gestation and on d 21 of lactation, mammary biopsies were performed on 10 sows per treatment to obtain parenchymal tissue samples for determination of mRNA abundance for , , , , , and genes. Milk samples and jugular blood samples were also obtained from sows on d 21 of lactation. Standard composition analyses (DM, fat, protein, and lactose) were done in milk. Concentrations of prolactin, IGF-1, glucose, and urea were measured in blood. There was a tendency for 21D sows to consume more feed than 2D or 7D sows during the first week of lactation ( < 0.10). There was no treatment effect on BW of piglets at any time until d 56 ( > 0.10). Concentrations of prolactin, IGF-1, urea, and glucose in sows on d 21 of lactation were not affected by treatment ( > 0.10). Dry matter, fat, protein, and lactose contents in milk were not altered by treatment ( > 0.10). On d 110 of gestation, gene expression was greater ( = 0.05) in 21D sows than in 7D sows. On d 21 of lactation, gene expression of was greater ( = 0.05) and that of tended to be lower ( < 0.10) in 7D sows than in 2D sows. The mRNA abundance of also tended to be lower ( < 0.10) in 2D sows than in 7D sows. Results indicate that increasing the duration of lactation from 2 d to 7 d or to 21 d in first-parity sows did not improve growth rate of their piglets in the subsequent lactation. This suggests that suckling of a teat for 2 d during the first lactation is sufficient to ensure optimal mammary development.
Collapse
|
15
|
Prognosis of pregnancy-associated breast cancer. Breast Cancer Res Treat 2017; 163:417-421. [DOI: 10.1007/s10549-017-4224-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/24/2017] [Indexed: 10/19/2022]
|
16
|
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: 52] [Impact Index Per Article: 7.4] [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
|
17
|
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: 15] [Impact Index Per Article: 1.9] [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
|
18
|
Sharp JA, Lefèvre C, Watt A, Nicholas KR. Analysis of human breast milk cells: gene expression profiles during pregnancy, lactation, involution, and mastitic infection. Funct Integr Genomics 2016; 16:297-321. [PMID: 26909879 DOI: 10.1007/s10142-016-0485-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 02/04/2016] [Accepted: 02/09/2016] [Indexed: 12/22/2022]
Abstract
The molecular processes underlying human milk production and the effects of mastitic infection are largely unknown because of limitations in obtaining tissue samples. Determination of gene expression in normal lactating women would be a significant step toward understanding why some women display poor lactation outcomes. Here, we demonstrate the utility of RNA obtained directly from human milk cells to detect mammary epithelial cell (MEC)-specific gene expression. Milk cell RNA was collected from five time points (24 h prepartum during the colostrum period, midlactation, two involutions, and during a bout of mastitis) in addition to an involution series comprising three time points. Gene expression profiles were determined by use of human Affymetrix arrays. Milk cells collected during milk production showed that the most highly expressed genes were involved in milk synthesis (e.g., CEL, OLAH, FOLR1, BTN1A1, and ARG2), while milk cells collected during involution showed a significant downregulation of milk synthesis genes and activation of involution associated genes (e.g., STAT3, NF-kB, IRF5, and IRF7). Milk cells collected during mastitic infection revealed regulation of a unique set of genes specific to this disease state, while maintaining regulation of milk synthesis genes. Use of conventional epithelial cell markers was used to determine the population of MECs within each sample. This paper is the first to describe the milk cell transcriptome across the human lactation cycle and during mastitic infection, providing valuable insight into gene expression of the human mammary gland.
Collapse
Affiliation(s)
- Julie A Sharp
- Institute for Frontier Materials, Deakin University, Geelong, VIC, 3216, Australia. .,Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia.
| | - Christophe Lefèvre
- Division of Bioinformatics, Walter and Eliza Hall Medical Research Institute, Melbourne, 3000, Australia
| | - Ashalyn Watt
- Institute for Frontier Materials, Deakin University, Geelong, VIC, 3216, Australia
| | - Kevin R Nicholas
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
19
|
Hassiotou F, Geddes DT. Immune cell-mediated protection of the mammary gland and the infant during breastfeeding. Adv Nutr 2015; 6:267-75. [PMID: 25979492 PMCID: PMC4424778 DOI: 10.3945/an.114.007377] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Breastfeeding has been regarded first and foremost as a means of nutrition for infants, providing essential components for their unique growth and developmental requirements. However, breast milk is also rich in immunologic factors, highlighting its importance as a mediator of protection. In accordance with its evolutionary origin, the mammary gland offers via the breastfeeding route continuation of the maternal to infant immunologic support established in utero. At birth, the infant's immune system is immature, and although it was exposed to the maternal microbial flora during pregnancy, it experiences an abrupt change in its microbial environment during and after birth, which is challenging and renders the infant highly susceptible to infection. Active and passive immunity protects the infant via breast milk, which is rich in immunoglobulins, lactoferrin, lysozyme, cytokines, and numerous other immunologic factors, including maternal leukocytes. Breast milk leukocytes provide active immunity and promote development of immunocompetence in the infant. Additionally, it has been speculated that they play a role in the protection of the mammary gland from infection. Leukocytes are thought to exert these functions via phagocytosis, secretion of antimicrobial factors and/or antigen presentation in both the mammary gland and the gastrointestinal tract of the infant, and also in other infant tissues, where they are transported via the systemic circulation. Recently, it has been demonstrated that breast milk leukocytes respond dynamically to maternal as well as infant infections, and are fewer in nonexclusively compared with exclusively breastfeeding dyads, further emphasizing their importance for both the mother and infant. This review summarizes the current knowledge of human milk leukocytes and factors influencing them, and presents recent novel findings supporting their potential as a diagnostic marker for infections of the lactating breast and of the breastfed infant.
Collapse
Affiliation(s)
- Foteini Hassiotou
- School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Crawley, Australia
| | | |
Collapse
|
20
|
Yasueda T, Oshima K, Nakatani H, Tabuchi K, Nadano D, Matsuda T. A protective effect of milk fat globule EGF factor VIII (MFG-E8) on the spontaneous fusion of milk fat globules in breast milk. J Biochem 2015; 158:25-35. [DOI: 10.1093/jb/mvv016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 01/01/2015] [Indexed: 01/13/2023] Open
|
21
|
Ponchon B, Lacasse P, Silanikove N, Ollier S, Zhao X. Effects of intramammary infusions of casein hydrolysate, ethylene glycol-bis(β-aminoethyl ether)-N,N,N′,N′-tetraacetic acid, and lactose at drying-off on mammary gland involution. J Dairy Sci 2014; 97:779-88. [DOI: 10.3168/jds.2013-7062] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 10/30/2013] [Indexed: 11/19/2022]
|
22
|
Silanikove N, Merin U, Shapiro F, Leitner G. Early mammary gland metabolic and immune responses during natural-like and forceful drying-off in high-yielding dairy cows. J Dairy Sci 2013; 96:6400-11. [DOI: 10.3168/jds.2013-6740] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 07/01/2013] [Indexed: 11/19/2022]
|
23
|
Mohammad MA, Haymond MW. Regulation of lipid synthesis genes and milk fat production in human mammary epithelial cells during secretory activation. Am J Physiol Endocrinol Metab 2013; 305:E700-16. [PMID: 23880316 PMCID: PMC3761196 DOI: 10.1152/ajpendo.00052.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Expression of genes for lipid biosynthetic enzymes during initiation of lactation in humans is unknown. Our goal was to study mRNA expression of lipid metabolic enzymes in human mammary epithelial cell (MEC) in conjunction with the measurement of milk fatty acid (FA) composition during secretory activation. Gene expression from mRNA isolated from milk fat globule (MFG) and milk FA composition were measured from 6 h to 42 days postpartum in seven normal women. Over the first 96 h postpartum, daily milk fat output increased severalfold and mirrored expression of genes for all aspects of lipid metabolism and milk FA production, including lipolysis at the MEC membrane, FA uptake from blood, intracellular FA transport, de novo FA synthesis, FA and glycerol activation, FA elongation, FA desaturation, triglyceride synthesis, cholesterol synthesis, and lipid droplet formation. Expression of the gene for a key lipid synthesis regulator, sterol regulatory element-binding transcription factor 1 (SREBF1), increased 2.0-fold by 36 h and remained elevated over the study duration. Expression of genes for estrogen receptor 1, thyroid hormone-responsive protein, and insulin-induced 2 increased progressively to plateau by 96 h. In contrast, mRNA of peroxisome proliferator-activated receptor-γ decreased severalfold. With onset of lactation, increased de novo synthesis of FA was the most prominent change in milk FA composition and mirrored the expression of FA synthesis genes. In conclusion, milk lipid synthesis and secretion in humans is a complex process requiring the orchestration of a wide variety of pathways of which SREBF1 may play a primary role.
Collapse
Affiliation(s)
- Mahmoud A Mohammad
- Department of Pediatrics, Children's Nutrition Research Center, U.S. Department of Agriculture/Agricultural Research Service, Baylor College of Medicine, Houston, Texas
| | | |
Collapse
|
24
|
Nakatani H, Yasueda T, Oshima K, Okajima T, Nadano D, Flint DJ, Matsuda T. Post-weaning increases in the milk-fat globule EGF-factor VIII on fat globules in mouse milk and in the uptake of the fat globules by HC11 mammary epithelial cells. J Biochem 2012; 153:31-41. [PMID: 23038672 DOI: 10.1093/jb/mvs116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Milk fat globules (MFGs) secreted by lactating mammary gland are unique lipid surrounded by a phospholipid bi-layer. We report here post-weaning changes in MFG EGF factor VIII (MFG-E8) and annexin V-accessible phosphatidyl-l-serine on the surface of MFGs. The MFG content in milk markedly decreased to about one-half within 2 days after forced weaning, despite a slight increase in milk protein content. Immunofluorescence-staining of MFGs using anti-MFG-E8 and annexin V indicated that MFG-E8 was present on some, but not all, MFGs before weaning, whereas most of MFGs were MFG-E8-positive and annexin V-negative after weaning. Free MFG-E8 with binding activity to phosphatidyl-l-serine was present abundantly in the post-weaning milk, and indeed exhibited binding to MFGs in pre-weaning milk. MFGs were taken up by HC11 mouse mammary epithelial cells in vitro, and those from post-weaning milk were remarkable for such cellular uptake. Moreover, the uptake of MFGs by the cells was inhibited by an anti-MFG-E8 antibody. Taken together, these findings suggest that MFG-E8 plays a critical role in regulation of MFG dynamics after weaning or during the suckling interval through the control of MFG-epithelial cell interaction in lactating mammary glands.
Collapse
Affiliation(s)
- Hajime Nakatani
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
Hassiotou F, Geddes D. Anatomy of the human mammary gland: Current status of knowledge. Clin Anat 2012; 26:29-48. [PMID: 22997014 DOI: 10.1002/ca.22165] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 08/16/2012] [Indexed: 12/18/2022]
Abstract
Mammary glands are unique to mammals, with the specific function of synthesizing, secreting, and delivering milk to the newborn. Given this function, it is only during a pregnancy/lactation cycle that the gland reaches a mature developmental state via hormonal influences at the cellular level that effect drastic modifications in the micro- and macro-anatomy of the gland, resulting in remodeling of the gland into a milk-secretory organ. Pubertal and post-pubertal development of the breast in females aids in preparing it to assume a functional state during pregnancy and lactation. Remarkably, this organ has the capacity to regress to a resting state upon cessation of lactation, and then undergo the same cycle of expansion and regression again in subsequent pregnancies during reproductive life. This plasticity suggests tight hormonal regulation, which is paramount for the normal function of the gland. This review presents the current status of knowledge of the normal macro- and micro-anatomy of the human mammary gland and the distinct changes it undergoes during the key developmental stages that characterize it, from embryonic life through to post-menopausal age. In addition, it discusses recent advances in our understanding of the normal function of the breast during lactation, with special reference to breastmilk, its composition, and how it can be utilized as a tool to advance knowledge on normal and aberrant breast development and function. Finally, anatomical and molecular traits associated with aberrant expansion of the breast are discussed to set the basis for future comparisons that may illuminate the origin of breast cancer.
Collapse
Affiliation(s)
- Foteini Hassiotou
- Hartmann Human Lactation Research Group, School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Crawley, Western Australia, Australia
| | | |
Collapse
|
26
|
Leitner G, Merin U, Krifucks O, Blum S, Rivas AL, Silanikove N. Effects of intra-mammary bacterial infection with coagulase negative staphylococci and stage of lactation on shedding of epithelial cells and infiltration of leukocytes into milk: Comparison among cows, goats and sheep. Vet Immunol Immunopathol 2012; 147:202-10. [DOI: 10.1016/j.vetimm.2012.04.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 04/16/2012] [Accepted: 04/18/2012] [Indexed: 11/29/2022]
|
27
|
Baumgarten SC, Frasor J. Minireview: Inflammation: an instigator of more aggressive estrogen receptor (ER) positive breast cancers. Mol Endocrinol 2012; 26:360-71. [PMID: 22301780 DOI: 10.1210/me.2011-1302] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Approximately 75% of breast tumors express the estrogen receptor (ER), and women with these tumors will receive endocrine therapy. Unfortunately, up to 50% of these patients will fail ER-targeted therapies due to either de novo or acquired resistance. ER-positive tumors can be classified based on gene expression profiles into Luminal A- and Luminal B-intrinsic subtypes, with distinctly different responses to endocrine therapy and overall patient outcome. However, the underlying biology causing this tumor heterogeneity has yet to become clear. This review will explore the role of inflammation as a risk factor in breast cancer as well as a player in the development of more aggressive, therapy-resistant ER-positive breast cancers. First, breast cancer risk factors, such as obesity and mammary gland involution after pregnancy, which can foster an inflammatory microenvironment within the breast, will be described. Second, inflammatory components of the tumor microenvironment, including tumor-associated macrophages and proinflammatory cytokines, which can act on nearby breast cancer cells and modulate tumor phenotype, will be explored. Finally, activation of the nuclear factor κB (NF-κB) pathway and its cross talk with ER in the regulation of key genes in the promotion of more aggressive breast cancers will be reviewed. From these multiple lines of evidence, we propose that inflammation may promote more aggressive ER-positive tumors and that combination therapy targeting both inflammation and estrogen production or actions could benefit a significant portion of women whose ER-positive breast tumors fail to respond to endocrine therapy.
Collapse
Affiliation(s)
- Sarah C Baumgarten
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, IL 60612, USA
| | | |
Collapse
|
28
|
Proinflammatory cytokines and CD14 expression in mammary tissue of cows following intramammary inoculation of Panax ginseng at drying off. Vet Immunol Immunopathol 2011; 144:52-60. [DOI: 10.1016/j.vetimm.2011.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 07/05/2011] [Accepted: 07/08/2011] [Indexed: 01/29/2023]
|
29
|
TIMP3 regulates mammary epithelial apoptosis with immune cell recruitment through differential TNF dependence. PLoS One 2011; 6:e26718. [PMID: 22053204 PMCID: PMC3203873 DOI: 10.1371/journal.pone.0026718] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 10/03/2011] [Indexed: 01/14/2023] Open
Abstract
Post-lactation mammary involution is a homeostatic process requiring epithelial apoptosis and clearance. Given that the deficiency of the extracellular metalloproteinase inhibitor TIMP3 impacts epithelial apoptosis and heightens inflammatory response, we investigated whether TIMP3 regulates these distinct processes during the phases of mammary gland involution in the mouse. Here we show that TIMP3 deficiency leads to TNF dysregulation, earlier caspase activation and onset of mitochondrial apoptosis. This accelerated first phase of involution includes faster loss of initiating signals (STAT3 activation; TGFβ3) concurrent with immediate luminal deconstruction through E-cadherin fragmentation. Epithelial apoptosis is followed by accelerated adipogenesis and a greater macrophage and T-cell infiltration in Timp3(-/-) involuting glands. Crossing in Tnf deficiency abrogates caspase 3 activation, but heightens macrophage and T-cell influx into Timp3(-/-) glands. The data indicate that TIMP3 differentially impacts apoptosis and inflammatory cell influx, based on involvement of TNF, during the process of mammary involution. An understanding of the molecular factors and wound healing microenvironment of the postpartum mammary gland may have implications for understanding pregnancy-associated breast cancer risk.
Collapse
|
30
|
Reactive oxygen species initiate luminal but not basal cell death in cultured human mammary alveolar structures: a potential regulator of involution. Cell Death Dis 2011; 2:e189. [PMID: 21814287 PMCID: PMC3181416 DOI: 10.1038/cddis.2011.69] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Post-lactational involution of the mammary gland is initiated within days of weaning. Clearing of cells occurs by apoptosis of the milk-secreting luminal cells in the alveoli and through stromal tissue remodeling to return the gland almost completely to its pre-pregnant state. The pathways that specifically target involution of the luminal cells in the alveoli but not the basal and ductal cells are poorly understood. In this study we show in cultured human mammary alveolar structures that the involution process is initiated by fresh media withdrawal, and is characterized by cellular oxidative stress, expression of activated macrophage marker CD68 and finally complete clearing of the luminal but not basal epithelial layer. This process can be simulated by ectopic addition of reactive oxygen species (ROS) in cultures without media withdrawal. Cells isolated from post-involution alveoli were enriched for the CD49f+ mammary stem cell (MaSC) phenotype and were able to reproduce a complete alveolar structure in subcultures without any significant loss in viability. We propose that the ROS produced by accumulated milk breakdown post-weaning may be the mechanism underlying the selective involution of secretory alveolar luminal cells, and that our culture model represents an useful means to investigate this and other mechanisms further.
Collapse
|
31
|
Coussens LM, Pollard JW. Leukocytes in mammary development and cancer. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a003285. [PMID: 21123394 DOI: 10.1101/cshperspect.a003285] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Leukocytes, of both the innate and adaptive lineages, are normal cellular components of all tissues. These important cells not only are critical for regulating normal tissue homeostasis, but also are significant paracrine regulators of all physiologic and pathologic tissue repair processes. This article summarizes recent insights regarding the trophic roles of leukocytes at each stage of mammary gland development and during cancer development, with a focus on Murids and humans.
Collapse
Affiliation(s)
- Lisa M Coussens
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 94143, USA
| | | |
Collapse
|
32
|
Mani O, Körner M, Sorensen MT, Sejrsen K, Wotzkow C, Ontsouka CE, Friis RR, Bruckmaier RM, Albrecht C. Expression, localization, and functional model of cholesterol transporters in lactating and nonlactating mammary tissues of murine, bovine, and human origin. Am J Physiol Regul Integr Comp Physiol 2010; 299:R642-54. [DOI: 10.1152/ajpregu.00723.2009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Members of the ATP-binding cassette (ABC) transporters play a pivotal role in cellular lipid efflux. To identify candidate cholesterol transporters implicated in lipid homeostasis and mammary gland (MG) physiology, we compared expression and localization of ABCA1, ABCG1, and ABCA7 and their regulatory genes in mammary tissues of different species during the pregnancy-lactation cycle. Murine and bovine mammary glands (MGs) were investigated during different functional stages. The abundance of mRNAs was determined by quantitative RT-PCR. Furthermore, transporter proteins were localized in murine, bovine, and human MGs by immunohistochemistry. In the murine MG, ABCA1 mRNA abundance was elevated during nonlactating compared with lactating stages, whereas ABCA7 and ABCA1 mRNA profiles were not altered. In the bovine MG, ABCA1, ABCG1, and ABCA7 mRNAs abundances were increased during nonlactating stages compared with lactation. Furthermore, associations between mRNA levels of transporters and their regulatory genes LXRα, PPARγ, and SREBPs were found. ABCA1, ABCG1, and ABCA7 proteins were localized in glandular MG epithelial cells (MEC) during lactation, whereas during nonlactating stages, depending on species, the proteins showed distinct localization patterns in MEC and adipocytes. Our results demonstrate that ABCA1, ABCG1, and ABCA7 are differentially expressed between lactation and nonlactating stages and in association with regulatory genes. Combined expression and localization data suggest that the selected cholesterol transporters are universal MG transporters involved in transport and storage of cholesterol and in lipid homeostasis of MEC. Because of the species-specific expression patterns of transporters in mammary tissue, mechanisms of cholesterol homeostasis seem to be differentially regulated between species.
Collapse
Affiliation(s)
- Orlando Mani
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Meike Körner
- Institute of Pathology, University of Bern, Switzerland
| | - Martin T. Sorensen
- Department of Animal Health, Welfare and Nutrition, Aarhus University, Tjele, Denmark
| | - Kristen Sejrsen
- Department of Animal Health, Welfare and Nutrition, Aarhus University, Tjele, Denmark
| | - Carlos Wotzkow
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Corneille E. Ontsouka
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Robert R. Friis
- Department of Clinical Research, University of Bern, Bern, Switzerland; and
| | | | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
33
|
Huser CA, Pringle MA, Heath VJ, Bell AK, Kendrick H, Smalley MJ, Crighton D, Ryan KM, Gusterson BA, Stein T. TSC-22D1 isoforms have opposing roles in mammary epithelial cell survival. Cell Death Differ 2009; 17:304-15. [PMID: 19745830 DOI: 10.1038/cdd.2009.126] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Transforming growth factor beta (TGFbeta)-stimulated clone-22 domain family member 1 (TSC-22D1) has previously been associated with enhanced apoptosis in several cell systems. In an attempt to identify novel factors that are involved in the control of cell death during mammary gland involution, we found that the mRNA for isoform 2 of TSC-22D1 was highly upregulated 24 h after forced weaning, when a dramatic increase in cell death occurred, closely following the expression of the known inducer of cell death during involution, TGFbeta3. This was paralleled by strongly increased TSC-22D1 isoform 2 protein levels in the luminal epithelium. In contrast, RNA and protein expression levels of the isoform 1 of TSC-22D1 did not change during development. Whereas isoform 2 induced cell death, isoform 1 suppressed TGFbeta-induced cell death and enhanced proliferation in mammary epithelial cell lines. Furthermore, four distinct forms of isoform 2 protein were detected in the mammary gland, of which only a 15-kDa form was associated with early involution. Our data describe novel opposing functions of the two mammalian TSC-22D1 isoforms in cell survival and proliferation, and establish the TSC-22D1 isoform 2 as a potential regulator of cell death during mammary gland involution.
Collapse
Affiliation(s)
- C A Huser
- Division of Cancer Sciences and Molecular Pathology, University of Glasgow, Glasgow, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mani O, Sorensen M, Sejrsen K, Bruckmaier R, Albrecht C. Differential expression and localization of lipid transporters in the bovine mammary gland during the pregnancy-lactation cycle. J Dairy Sci 2009; 92:3744-56. [DOI: 10.3168/jds.2009-2063] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
35
|
Stein T, Salomonis N, Nuyten DSA, van de Vijver MJ, Gusterson BA. A mouse mammary gland involution mRNA signature identifies biological pathways potentially associated with breast cancer metastasis. J Mammary Gland Biol Neoplasia 2009; 14:99-116. [PMID: 19408105 DOI: 10.1007/s10911-009-9120-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Accepted: 04/06/2009] [Indexed: 12/15/2022] Open
Abstract
Mouse mammary gland involution resembles a wound healing response with suppressed inflammation. Wound healing and inflammation are also associated with tumour development, and a 'wound-healing' gene expression signature can predict metastasis formation and survival. Recent studies have shown that an involuting mammary gland stroma can promote metastasis. It could therefore be hypothesised that gene expression signatures from an involuting mouse mammary gland may provide new insights into the physiological pathways that promote breast cancer progression. Indeed, using the HOPACH clustering method, the human orthologues of genes that were differentially regulated at day 3 of mammary gland involution and showed prolonged expression throughout the first 4 days of involution distinguished breast cancers in the NKI 295 breast cancer dataset with low and high metastatic activity. Most strikingly, genes associated with copper ion homeostasis and with HIF-1 promoter binding sites were the most over-represented, linking this signature to hypoxia. Further, six out of the ten mRNAs with strongest up-regulation in cancers with poor survival code for secreted factors, identifying potential candidates that may be involved in stromal/matrix-enhanced metastasis formation/breast cancer development. This method therefore identified biological processes that occur during mammary gland involution, which may be critical in promoting breast cancer metastasis that could form a basis for future investigation, and supports a role for copper in breast cancer development.
Collapse
Affiliation(s)
- Torsten Stein
- Division of Cancer Sciences and Molecular Pathology, Section of Gene Regulation and Mechanisms of Disease, Western Infirmary, University of Glasgow, Glasgow, UK.
| | | | | | | | | |
Collapse
|
36
|
Flanders KC, Wakefield LM. Transforming growth factor-(beta)s and mammary gland involution; functional roles and implications for cancer progression. J Mammary Gland Biol Neoplasia 2009; 14:131-44. [PMID: 19396528 PMCID: PMC2797127 DOI: 10.1007/s10911-009-9122-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Accepted: 04/15/2009] [Indexed: 01/07/2023] Open
Abstract
During rodent mammary gland involution there is a dramatic increase in the expression of the transforming growth factor-beta isoform, TGF-beta3. The TGF-betas are multifunctional cytokines which play important roles in wound healing and in carcinogenesis. The responses that are activated in the remodeling of the gland during involution have many similarities with the wound healing process and have been postulated to generate a mammary stroma that provides a microenvironment favoring tumor progression. In this review we will discuss the putative role of TGF-beta during involution, as well as its effects on the mammary microenvironment and possible implications for pregnancy-associated tumorigenesis.
Collapse
Affiliation(s)
- Kathleen C Flanders
- Laboratory of Cancer Biology & Genetics, National Cancer Institute, Bethesda, MD 20892, USA.
| | | |
Collapse
|
37
|
Kumar Y, Chahal A, Garg M, Bhatia A, Mahajan NC, Ganju A. Delayed involution of lactation presenting as a non-resolving breast mass: a case report. J Med Case Rep 2008; 2:327. [PMID: 18851738 PMCID: PMC2572069 DOI: 10.1186/1752-1947-2-327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Accepted: 10/13/2008] [Indexed: 11/10/2022] Open
Abstract
Introduction Involution of lactation is a physiological process. Rarely, it may be delayed and troublesome for the lactating woman. Though lactation-induced changes in breast are well known, morphological features of delayed involution are not clear. Case presentation We report a case of a 22-year-old lactating mother who presented with a painful, non-resolving breast mass 5 months after delivery. Clinically, it simulated an inflammatory carcinoma. Histopathology, however, revealed involuting lactational changes. Conclusion To the best of our knowledge, lactational involution with such a presentation has not been described in the English literature. The case needs to be reported so that this entity can be considered among the differential diagnoses of breast masses in a lactating patient.
Collapse
Affiliation(s)
- Yashwant Kumar
- Department of Pathology, Maharshi Markandeshwar Institute of Medical Sciences and Research, Mullana, Ambala Haryana, India.
| | | | | | | | | | | |
Collapse
|
38
|
Differential expression of ABC transporters and their regulatory genes during lactation and dry period in bovine mammary tissue. J DAIRY RES 2008; 75:406-14. [PMID: 18700997 DOI: 10.1017/s002202990800335x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
ATP-binding cassette (ABC) transporters play a pivotal role in human physiology, and mutations in these genes often result in severe hereditary diseases. ABC transporters are expressed in the bovine mammary gland but their physiological role in this organ remains elusive. Based on findings in the context of human disorders we speculated that candidate ABC transporters are implicated in lipid and cholesterol transport in the mammary gland. Therefore we investigated the expression pattern of selected genes that are associated with sterol transport in lactating and nonlactating mammary glands of dairy cows. mRNA levels from mammary gland biopsies taken during lactation and in the first and second week of the dry period were analysed using quantitative PCR. Five ABC transporter genes, namely ABCA1, ABCA7, ABCG1, ABCG2 and ABCG5, their regulating genes LXRalpha, PPARgamma, SREBP1 and the milk proteins lactoferrin and alpha-lactalbumin were assessed. A significantly enhanced expression in the dry period was observed for ABCA1 while a significant decrease of expression in this period was detected for ABCA7, ABCG2, SREBP1 and alpha-lactalbumin. ABCG1, ABCG5, LXRalpha, PPARgamma and lactoferrin expression was not altered between lactation and dry period. These results indicate that candidate ABC transporters involved in lipid and cholesterol transport show differential mRNA expression between lactation and the dry period. This may be due to physiological changes in the mammary gland such as immigration of macrophages or the accumulation of fat due to the loss of liquid in the involuting mammary gland. The current mRNA expression analysis of transporters in the mammary gland is the prerequisite for elucidating novel molecular mechanisms underlying cholesterol and lipid transfer into milk.
Collapse
|
39
|
Hojilla CV, Wood GA, Khokha R. Inflammation and breast cancer: metalloproteinases as common effectors of inflammation and extracellular matrix breakdown in breast cancer. Breast Cancer Res 2008; 10:205. [PMID: 18394187 PMCID: PMC2397522 DOI: 10.1186/bcr1980] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Two rapidly evolving fields are converging to impact breast cancer: one has identified novel substrates of metalloproteinases that alter immune cell function, and the other has revealed a role for inflammation in human cancers. Evidence now shows that the mechanisms underlying these two fields interact in the context of breast cancer, providing new opportunities to understand this disease and uncover novel therapeutic strategies. The metalloproteinase class of enzymes is well studied in mammary gland development and physiology, but mostly in the context of extracellular matrix modification. Aberrant metalloproteinase expression has also been implicated in breast cancer progression, where these genes act as tumor modifiers. Here, we review how the metalloproteinase axis impacts mammary physiology and tumorigenesis and is associated with inflammatory cell influx in human breast cancer, and evaluate its potential as a regulator of inflammation in the mammary gland.
Collapse
Affiliation(s)
- Carlo V Hojilla
- Department of Medical Biophysics, Ontario Cancer Institute, Toronto, M5G 2M9 Canada
| | | | | |
Collapse
|
40
|
Monks J, Smith-Steinhart C, Kruk ER, Fadok VA, Henson PM. Epithelial cells remove apoptotic epithelial cells during post-lactation involution of the mouse mammary gland. Biol Reprod 2007; 78:586-94. [PMID: 18057312 DOI: 10.1095/biolreprod.107.065045] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Following the cessation of lactation, the mammary gland undergoes a physiologic process of tissue remodeling called involution in which glandular structures are lost, leaving an adipose tissue compartment that takes up a much larger proportion of the tissue. A quantitative morphometric analysis was undertaken to determine the mechanisms for clearance of the epithelial cells during this process. The involution process was set in motion by removal of pups from 14-day lactating C57BL/6 mice. Within hours, milk-secreting epithelial cells were shed into the glandular lumen. These cells became apoptotic, exhibiting exposure of phosphatidylserine residues on their surfaces, activation of effector caspase-3, staining for caspase-cleaved keratin 18, loss of internal organellar structure, and nuclear breakdown, but minimal blebbing or generation of apoptotic bodies. Clearance of residual milk and the shed epithelial cells was rapid, with most of the removal occurring in the first 72 h. Intact apoptotic epithelial cells were engulfed in large numbers by residual viable epithelial cells into spacious efferosomes. This process led to essentially complete involution within 4 days, at which point estrous cycling recommenced. Macrophages and other inflammatory cells did not contribute to the clearance of either residual milk or apoptotic cells, which appeared to be due entirely to the epithelium itself.
Collapse
Affiliation(s)
- Jenifer Monks
- Program in Cell Biology, Department of Pediatrics, National Jewish Medical & Research Center, Denver, Colorado 80206, USA.
| | | | | | | | | |
Collapse
|
41
|
Fiaschi M, Rozell B, Bergström Å, Toftgård R, Kleman MI. Targeted Expression of GLI1 in the Mammary Gland Disrupts Pregnancy-induced Maturation and Causes Lactation Failure. J Biol Chem 2007; 282:36090-101. [DOI: 10.1074/jbc.m704280200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
42
|
Abstract
Mastitis, an inflammatory reaction of the mammary gland that is usually caused by a microbial infection, is recognized as the most costly disease in dairy cattle. Decreased milk production accounts for approximately 70% of the total cost of mastitis. Mammary tissue damage reduces the number and activity of epithelial cells and consequently contributes to decreased milk production. Mammary tissue damage has been shown to be induced by either apoptosis or necrosis. These 2 distinct types of cell death can be distinguished by morphological, biochemical, and molecular changes in dying cells. Both bacterial factors and host immune reactions contribute to epithelial tissue damage. During infection of the mammary glands, the tissue damage can initially be caused by bacteria and their products. Certain bacteria produce toxins that destroy cell membranes and damage milk-producing tissue, whereas other bacteria are able to invade and multiply within the bovine mammary epithelial cells before causing cell death. In addition, mastitis is characterized by an influx of somatic cells, primarily polymorphonuclear neutrophils, into the mammary gland. With more immune cells migrating into the mammary gland and the breakdown of the blood-milk barrier, damage to the mammary epithelium worsens. It is well known that breakdown of the extracellular matrix can lead to death of the epithelial cells. Meanwhile, polymorphonuclear neutrophils can harm the mammary tissue by releasing reactive oxygen intermediates and proteolytic enzymes. In vitro and in vivo studies suggest that the use of antioxidants and other protective compounds in mastitis control programs is worth investigating, because they may aid in alleviating damage to secretory cells and thus reduce subsequent milk loss.
Collapse
Affiliation(s)
- X Zhao
- Department of Animal Science, McGill University, Ste. Anne de Bellevue, Québec, H9X 3V9, Canada.
| | | |
Collapse
|
43
|
Lee S, Medina D, Tsimelzon A, Mohsin SK, Mao S, Wu Y, Allred DC. Alterations of gene expression in the development of early hyperplastic precursors of breast cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:252-62. [PMID: 17591970 PMCID: PMC1941596 DOI: 10.2353/ajpath.2007.061010] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Enlargement of normal terminal duct lobular units (TDLUs) by hyperplastic columnar epithelial cells is one of the most common abnormalities of growth in the adult female human breast. These hyperplastic enlarged lobular units (HELUs) are important clinically as the earliest histologically identifiable potential precursor of breast cancer. The causes of the hyperplasia are unknown but may include estrogen-simulated growth mediated by estrogen receptor-alpha, which is highly elevated in HELUs and may be fundamental to their development. The present study used DNA microarray technology and RNA from microdissected pure epithelial cells to examine changes in gene expression and molecular pathways associated with the development of HELUs from TDLUs. The results suggest that HELUs evolve from TDLUs primarily by reactivation of pathways involved in embryonic development and suppression of terminal differentiation. Changes in ERBB genes were particularly prominent, including a uniform switch in ligands for the ERBB1 receptor (14-fold decrease in epidermal growth factor and 10-fold increase in amphiregulin, respectively) in HELUs compared with TDLUs. Epidermal growth factor regulates terminal differentiation in adult breast and amphiregulin is critical to normal embryonic breast development. Because HELUs are such early potential precursors of breast cancer, targeting some of these alterations may be especially promising strategies for breast cancer prevention.
Collapse
Affiliation(s)
- Sangjun Lee
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Ning Y, Hoang B, Schuller AGP, Cominski TP, Hsu MS, Wood TL, Pintar JE. Delayed mammary gland involution in mice with mutation of the insulin-like growth factor binding protein 5 gene. Endocrinology 2007; 148:2138-47. [PMID: 17255210 DOI: 10.1210/en.2006-0041] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
IGFs (IGF-I and IGF-II) are essential for development, and their bioactivities are tightly regulated by six related IGF-binding proteins (IGFBPs). IGFBP-5 is the most highly conserved binding protein and is expressed in several key developmental lineages as well as in multiple adult tissues including the mammary gland. To explore IGFBP-5 actions in vivo, we produced IGFBP-5 knockout (KO) mice. Whole-body growth, selected organ weights, and body composition were essentially normal in IGFBP-5 KO mice, presumably because of substantial compensation by remaining IGFBP family members. The IGFBP-5 KO mice also exhibited normal mammary gland development and were capable of nursing their pups. We then directly evaluated the proposed role of IGFBP-5 in apoptosis and remodeling of mammary gland during involution. We found that the process of involution after forced weaning was delayed in IGFBP-5 KO mice, with both the appearance of apoptotic cells and the reappearance of adipocytes retarded in mutant mice, compared with controls. We also determined the effects of IGFBP-5 deletion on mammary gland development in pubertal females after ovariectomy and stimulation with estradiol/progesterone. In this paradigm, IGFBP-5 KO mammary glands exhibited enhanced alveolar bud formation consistent with enhanced IGF-I action. These results demonstrate that IGFBP-5, although not essential for normal growth, is required for normal mammary gland involution and can regulate mammary gland morphogenesis in response to hormone stimulation.
Collapse
Affiliation(s)
- Yun Ning
- Department of Neuroscience, University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Russell JS, McGee SO, Ip MM, Kuhlmann D, Masso-Welch PA. Conjugated linoleic acid induces mast cell recruitment during mouse mammary gland stromal remodeling. J Nutr 2007; 137:1200-7. [PMID: 17449582 PMCID: PMC2447169 DOI: 10.1093/jn/137.5.1200] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Conjugated linoleic acid (CLA) is a dietary chemopreventive agent that induces apoptosis in the mammary adipose vascular endothelium and decreases mammary brown adipose tissue (BAT) and white adipose tissue (WAT). To determine onset and extent of stromal remodeling, we fed CD2F1/Cr mice diets supplemented with 1 or 2 g/100 g mixed CLA isomers for 1-7 wk. BAT loss, collagen deposition, and leukocyte recruitment occurred in the mouse mammary fat pad, coincident with an increase in parenchymal-associated mast cells in mice fed both levels of CLA. Feeding experiments with purified isomers (0.5 g/100 g diet) demonstrated that these changes were induced by trans-10, cis-12 CLA (10,12-CLA), but not by cis-9, trans-11 CLA (9,11-CLA). This stromal remodeling did not require tumor necrosis factor (TNF)-alpha, a major cytokine in mast cells, as TNF-alpha null mice demonstrated collagen deposition, increased leukocytes, and BAT loss in the mammary fat pad in response to 10,12-CLA. To test the hypothesis that mast cells recruited in response to 10,12-CLA were required for stromal remodeling, Steel mice (WBB6F1/J-kit(W)/kit(W-V)), which lack functional mast cells, were examined for their stromal response to 10,12-CLA. Both wild-type and Steel mice showed a significantly increased leukocytic adipose infiltrate, collagen deposition, and decreased adipocyte size, although BAT was maintained in Steel mice. These results demonstrate that 10,12-CLA induces an inflammatory and fibrotic phenotype in the mouse mammary gland stroma that is independent of TNF-alpha or mast cells and suggest caution in the use of 10,12-CLA for breast cancer chemoprevention.
Collapse
Affiliation(s)
- Joshua S. Russell
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Sibel Oflazoglu McGee
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Margot M. Ip
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Dietrich Kuhlmann
- Department of Mathematics and Statistics, Canisius College, Buffalo, NY 14208
| | - Patricia A. Masso-Welch
- Department of Biotechnical and Clinical Laboratory Sciences, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14214
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
46
|
Sharp JA, Lefevre C, Brennan AJ, Nicholas KR. The fur seal-a model lactation phenotype to explore molecular factors involved in the initiation of apoptosis at involution. J Mammary Gland Biol Neoplasia 2007; 12:47-58. [PMID: 17431798 DOI: 10.1007/s10911-007-9037-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Mammary gland involution requires co-ordination of milk production, immune responses, apoptosis and remodeling. Initiation and progression of each of these components involves integral control by the mammary gland. Although cell-based culture models and genetically manipulated animals have shed light on these processes, the factors controlling each step in the involution cascade are still poorly understood. The fur seal displays a unique lactation phenotype. During the lactation cycle the mammary gland downregulates milk production and initiates an immune response but fails to initiate the apoptotic phase of involution, allowing the female fur seal to undertake long foraging trips of up to 28 days between suckling bouts. Upon return to shore the female continues feeding her pup following resumption of lactation and milk production. Expression profiling of genes involved in this lactation cycle provides valuable tools for investigation of the factors responsible for the initiation of apoptosis at involution.
Collapse
Affiliation(s)
- Julie A Sharp
- CRC for Innovative Dairy Products, Department of Zoology, University of Melbourne, Melbourne, Victoria 3010, Australia.
| | | | | | | |
Collapse
|
47
|
Schedin P, O'Brien J, Rudolph M, Stein T, Borges V. Microenvironment of the involuting mammary gland mediates mammary cancer progression. J Mammary Gland Biol Neoplasia 2007; 12:71-82. [PMID: 17318269 DOI: 10.1007/s10911-007-9039-3] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Breast cancer diagnosed after a completed pregnancy has higher metastatic potential and therefore a much poorer prognosis. We hypothesize that following pregnancy the process of mammary gland involution, which returns the gland to its pre-pregnant state, co-opts some of the programs of wound healing. The pro-inflammatory milieu that results, while physiologically normal, promotes tumor progression. In this review, the similarities between mammary gland involution after cessation of milk-production and pathological tissue remodeling are discussed in light of emerging data demonstrating a role for pathological tissue remodeling in cancer.
Collapse
Affiliation(s)
- Pepper Schedin
- AMC Cancer Research Center, University of Colorado Health Science Center, Aurora, CO, USA.
| | | | | | | | | |
Collapse
|
48
|
Ollier S, Robert-Granié C, Bernard L, Chilliard Y, Leroux C. Mammary transcriptome analysis of food-deprived lactating goats highlights genes involved in milk secretion and programmed cell death. J Nutr 2007; 137:560-7. [PMID: 17311940 DOI: 10.1093/jn/137.3.560] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Animal nutrition considerably affects milk composition that influences its nutritional quality. Milk component synthesis and secretion by the mammary gland involve expression of a large number of genes whose nutritional regulation remains poorly defined. In this study, we examined the effect of food deprivation (FD) on the expression of 8379 genes in caprine mammary gland using a bovine oligonucleotide microarray. Twelve lactating goats were assigned to 2 groups based on their feeding level (control diet ad libitum vs. 48-h FD). We identified 161 genes whose expression was altered by FD. Most of these genes (88%) were downregulated, suggesting a stress response by the mammary gland. In particular, the decrease in expression of genes involved in milk protein, lactose, and lipid metabolism could contribute together with the shortage of nutrients to the drop in milk protein, lactose, and fat secretion. In addition, this study highlights modification of the expression of at least 14 genes that could be responsible for a slowdown in mammary cell proliferation and differentiation and/or an increase in programmed cell death in response to 48-h FD in goats.
Collapse
Affiliation(s)
- Séverine Ollier
- Unité de Recherches sur les Herbivores, Institut National de la Recherche Agronomique, Theix, 63122 Saint Genès-Champanelle, France
| | | | | | | | | |
Collapse
|
49
|
Atabai K, Sheppard D, Werb Z. Roles of the innate immune system in mammary gland remodeling during involution. J Mammary Gland Biol Neoplasia 2007; 12:37-45. [PMID: 17286210 PMCID: PMC2574498 DOI: 10.1007/s10911-007-9036-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mammary gland involution is a period of intensive tissue remodeling. Over the course of a relatively brief period, a large proportion of the mammary gland epithelium undergoes programmed cell death and is removed by phagocytes. In addition, the gland is cleared of residual milk fat globules as well as milk and adipocytes become the predominant cell type. The role of the immune system in this process has not been clearly defined. Professional phagocytes derived from the immune system can participate in the clearance of apoptotic and autophagic cells, the removal of residual milk components, and the prevention of mastitis during mammary gland involution. However, many of these functions can also be performed by non-professional phagocytes (e.g. mammary epithelial cells). This review will discuss the evidence that supports a role for innate immune cells in mammary gland remodeling during involution.
Collapse
Affiliation(s)
- Kamran Atabai
- Lung Biology Center, Cardiovascular Research Institute, Department of Medicine, University of California, San Francisco, CA 94143-2922, USA.
| | | | | |
Collapse
|
50
|
Schwertfeger KL, Rosen JM, Cohen DA. Mammary gland macrophages: pleiotropic functions in mammary development. J Mammary Gland Biol Neoplasia 2006; 11:229-38. [PMID: 17115264 DOI: 10.1007/s10911-006-9028-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Mammary gland development is a complex process involving epithelial cells and supporting stromal cells. Macrophages (MØs) are an important component of the mammary gland stroma and are critical for normal mammary gland development; however, the mechanisms by which macrophages regulate these processes are not well understood. MØs are known to interact with numerous cell types, including epithelial cells, fibroblasts, adipocytes, and endothelial cells, all of which are significant components of mammary gland development. Therefore, the purpose of this review is to describe the interactions between MØs and these various cell types and use this knowledge to identify potential functions of MØs in the mammary gland.
Collapse
Affiliation(s)
- Kathryn L Schwertfeger
- Department of Lab Medicine and Pathology, University of Minnesota Cancer Center, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455, USA.
| | | | | |
Collapse
|