1
|
Danev N, Poggi JM, Dewever EA, Bartlett AP, Oliveira L, Huntimer L, Harman RM, Van de Walle GR. Immortalized mammosphere-derived epithelial cells retain a bioactive secretome with antimicrobial, regenerative, and immunomodulatory properties. Stem Cell Res Ther 2024; 15:429. [PMID: 39543714 PMCID: PMC11566417 DOI: 10.1186/s13287-024-04019-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND The secretome of primary bovine mammosphere-derived epithelial cells (MDECs) has been shown to exert antimicrobial, regenerative, and immunomodulatory properties in vitro, which warrants its study as a potential biologic treatment with the potential to be translated to human medicine. Currently, the use of the MDEC secretome as a therapy is constrained by the limited life span of primary cell cultures and the decrease of secretome potency over cell passages. METHODS To address these limitations, early-passage bovine MDECs were immortalized using hTERT, a human telomerase reverse transcriptase. The primary and immortal MDECs were compared morphologically, transcriptomically, and phenotypically. The functional properties and proteomic profiles of the secretome of both cell lines were evaluated and compared. All experiments were performed with both low and high passage cell cultures. RESULTS We confirmed through in vitro experiments that the secretome of immortalized MDECs, unlike that of primary cells, maintained antimicrobial and pro-migratory properties over passages, while pro-angiogenic effects of the secretome from both primary and immortalized MDECs were lost when the cells reached high passage. The secretome from primary and immortalized MDECs, at low and high passages exerted immunomodulatory effects on neutrophils in vitro. CONCLUSIONS High passage immortalized MDECs retain a bioactive secretome with antimicrobial, regenerative, and immunomodulatory properties, suggesting they may serve as a consistent cell source for therapeutic use.
Collapse
Affiliation(s)
- Nikola Danev
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Julia M Poggi
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Emilie A Dewever
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Arianna P Bartlett
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Leane Oliveira
- Elanco Animal Health, 2500 Innovation Way, Indianapolis, IN, 46241, USA
| | - Lucas Huntimer
- Elanco Animal Health, 2500 Innovation Way, Indianapolis, IN, 46241, USA
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA.
- Department of Veterinary Pathobiology, Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, UK.
| |
Collapse
|
2
|
Morato A, Accornero P, Hovey RC. ERBB Receptors and Their Ligands in the Developing Mammary Glands of Different Species: Fifteen Characters in Search of an Author. J Mammary Gland Biol Neoplasia 2023; 28:10. [PMID: 37219601 DOI: 10.1007/s10911-023-09538-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/26/2023] [Indexed: 05/24/2023] Open
Abstract
The ERBB tyrosine kinase receptors and their ligands belong to a complex family that has diverse biological effects and expression profiles in the developing mammary glands, where its members play an essential role in translating hormone signals into local effects. While our understanding of these processes stems mostly from mouse models, there is the potential for differences in how this family functions in the mammary glands of other species, particularly in light of their unique histomorphological features. Herein we review the postnatal distribution and function of ERBB receptors and their ligands in the mammary glands of rodents and humans, as well as for livestock and companion animals. Our analysis highlights the diverse biology for this family and its members across species, the regulation of their expression, and how their roles and functions might be modulated by varying stromal composition and hormone interactions. Given that ERBB receptors and their ligands have the potential to influence processes ranging from normal mammary development to diseased states such as cancer and/or mastitis, both in human and veterinary medicine, a more complete understanding of their biological functions should help to direct future research and the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Alessia Morato
- Department of Animal Science, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA.
| | - Paolo Accornero
- Department of Veterinary Science, University of Turin, Largo Paolo Braccini 2, Grugliasco, TO, 10095, Italy
| | - Russell C Hovey
- Department of Animal Science, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| |
Collapse
|
3
|
Altamirano GA, Masat E, Rivera O, Alarcón R, Dioguardi G, Muñoz-de-Toro M, Luque EH, Kass L. Postnatal exposure to a glyphosate-based herbicide interferes with the development and growth of the mammary gland of pre-pubertal Ewe lambs. CHEMOSPHERE 2023; 313:137358. [PMID: 36427587 DOI: 10.1016/j.chemosphere.2022.137358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/07/2022] [Accepted: 11/21/2022] [Indexed: 06/16/2023]
Abstract
The aim of the present study was to evaluate whether early postnatal exposure to a glyphosate-based herbicide (GBH) alters pre-pubertal mammary development in Friesian lambs. To this end, from postnatal day 1-14, ewe lambs were exposed subcutaneously or orally to GBH (2 mg/kg bw/day) or vehicle (control) and mammary gland biopsies were obtained at 45 days of age. GBH-exposed lambs exhibited larger mammary ducts and less area occupied by terminal duct lobular units than controls, accompanied by an increase in the area of adipocytes in the mammary stroma. Lambs subcutaneously exposed to GBH showed increased protein expression of estrogen receptor alpha; however, both GBH-exposed groups had decreased mRNA expression of this receptor. Control lambs showed nuclear progesterone receptor (PR) protein expression, whereas GBH-exposed animals showed cytoplasmic PR expression; both GBH-exposed groups exhibited decreased mRNA expression of PR. GBH-exposed lambs also had decreased epithelial cell proliferation. Regarding insulin-like growth factors, both groups showed similar IGF-1 mRNA and protein expression but decreased expression of its receptor, and increased IGFBP5 expression. In addition, phosphorylated AKT was only observed in the mammary gland of control lambs. Our results show that early postnatal exposure to GBH, regardless of the exposure route, affects the IGF-1 system and the AKT/protein kinase B pathway, interfering with steroid hormone receptor expression and cell proliferation. This consequently modifies the growth and development of the pre-pubertal mammary gland of Frisian lambs.
Collapse
Affiliation(s)
- Gabriela A Altamirano
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Eduardo Masat
- Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Oscar Rivera
- Instituto de Investigación Sobre Producción Agropecuaria, Ambiente y Salud (IIPAAs), Facultad de Ciencias Agrarias, Universidad Nacional de Lomas de Zamora, Buenos Aires, Argentina
| | - Ramiro Alarcón
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Gisela Dioguardi
- Instituto de Investigación Sobre Producción Agropecuaria, Ambiente y Salud (IIPAAs), Facultad de Ciencias Agrarias, Universidad Nacional de Lomas de Zamora, Buenos Aires, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Enrique H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Laura Kass
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
4
|
Dado-Senn BM, Field SL, Davidson BD, Dahl GE, Laporta J. In utero hyperthermia in late gestation derails dairy calf early-life mammary development. J Anim Sci 2022; 100:skac186. [PMID: 36206013 PMCID: PMC9541282 DOI: 10.1093/jas/skac186] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/23/2022] [Indexed: 11/06/2022] Open
Abstract
Prenatal hyperthermia has immediate and long-term consequences on dairy cattle growth, immunity, and productivity. While changes in the molecular architecture are reported in the mature mammary gland (MG), any influence on early-life mammary development is unknown. Herein, we characterize the impact of late-gestation in utero heat stress on heifer mammary gross and cellular morphology at early-life developmental stages (i.e., birth and weaning). During summer, pregnant dams were exposed to environmental heat stress (shade of a free-stall barn) or offered active cooling (shade, fans, and water soakers) for 54 ± 5 d before parturition (avg. temperature-humidity index = 79). Heifer calves born to these dams were either in utero heat-stressed (IU-HT; n = 36) or in utero cooled (IU-CL; n = 37) and were managed as a single cohort thereafter. A subset of heifers was euthanized at birth (d0; n = 8/treatment; 4.6 ± 2.3 h after birth) and after weaning (d63; n = 8/treatment; 63.0 ± 1.5 d) to harvest the whole MG. An ultrasound of rear mammary parenchyma (MPAR) was taken prior to d63 and correlated to harvested MPAR cross-sectional area and weight. Portions of mammary fat pad (MFP) and MPAR were preserved for compositional and histological analysis, including ductal structure number and cross-sectional area, connective tissue area, and adipocyte number and cross-sectional area. Cellular proliferation in MPAR was assessed via Ki-67 immunohistochemistry. Relative to IU-CL heifers, the MGs of IU-HT heifers were shorter in length at d0 and d63 (P ≤ 0.02). There were moderate correlations between d63 ultrasound and harvest measures. The IU-HT heifers had reduced MG and MFP mass at d0 and d63 (P ≤ 0.05), whereas MPAR mass was reduced only at d0 (P = 0.01). IU-HT heifers had greater MPAR protein and DNA content at d63 (P ≤ 0.04), but there were no MFP compositional differences (P ≥ 0.12). At d0, IU-HT heifers had fewer MPAR ductal structures (P ≤ 0.06), but there were no differences at d63. Yet, MPAR luminal and total ductal structure cross-sectional areas of IU-HT heifers were reduced at both d0 and d63 (P ≤ 0.01). The MFP adipocytes of IU-HT heifers were smaller at d0 (P ≤ 0.01), but differences were not detected at d63. The IU-HT heifers had diminished MPAR total, stromal, and epithelial cellular proliferation at both d0 and d63 (P < 0.01). Prenatal hyperthermia derails dairy calf early-life mammary development with potential carry-over consequences on future synthetic capacity.
Collapse
Affiliation(s)
- Bethany M Dado-Senn
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sena L Field
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Brittney D Davidson
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Geoffrey E Dahl
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Jimena Laporta
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
5
|
Aalam SMM, Beer PA, Kannan N. Assays for functionally defined normal and malignant mammary stem cells. Adv Cancer Res 2019; 141:129-174. [PMID: 30691682 DOI: 10.1016/bs.acr.2018.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The discovery of rare, heterogeneous self-renewing stem cells with shared developmental and molecular features within epithelial components of mammary gland and breast cancers has provided a conceptual framework to understand cellular composition of these tissues and mechanisms that control their number. These normal mammary epithelial stem cells (MaSCs) and breast cancer stem cells (BCSCs) were identified and analyzed using transplant assays (namely mammary repopulating unit (MRU) assay, mammary tumor-initiating cell (TIC) assay), which reveal their latent ability to regenerate respective normal and malignant epithelial tissues with self-renewing units displaying hierarchical cellular differentiation over multiple generations in recipient mice. "Next-generation" methods using "barcoded" normal and malignant mammary cells, with the help of next-generation sequencing (NGS) technology, have revealed hidden complexity and heterogeneous growth potential of MaSCs and BCSCs. Several single markers or combinations of markers have been reported to prospectively enrich MaSCs and BCSCs. Such markers and the extent to which they enrich for MaSCs and BCSCs activity require a critical appraisal. Also, knowledge of the functional assays and their limitations and harmonious reporting of results is a prerequisite to improve our understanding of MaSCs and BCSCs. This chapter describes evolution of the concept of MaSCs and BCSCs, and specific methodologies to investigate them.
Collapse
Affiliation(s)
- Syed Mohammed Musheer Aalam
- Laboratory of Stem Cell and Cancer Biology, Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Philip Anthony Beer
- Laboratory of Stem Cell and Cancer Biology, Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States; Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Nagarajan Kannan
- Laboratory of Stem Cell and Cancer Biology, Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
6
|
Hare KS, Leal LN, Romao JM, Hooiveld GJ, Soberon F, Berends H, Van Amburgh ME, Martín-Tereso J, Steele MA. Preweaning nutrient supply alters mammary gland transcriptome expression relating to morphology, lipid accumulation, DNA synthesis, and RNA expression in Holstein heifer calves. J Dairy Sci 2019; 102:2618-2630. [PMID: 30612800 DOI: 10.3168/jds.2018-15699] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 11/04/2018] [Indexed: 01/16/2023]
Abstract
The objective of this study was to analyze the mammary gland transcriptome to determine how preweaning nutrient supply alters the molecular mechanisms that regulate preweaning mammary development. Holstein heifers were fed via milk replacer (MR) either an elevated level of nutrient intake (ELE; on average, 5.9 ± 0.2 Mcal of ME in 8.4 L of MR/d, n = 6) or a restricted amount of nutrients (RES; 2.8 ± 0.2 Mcal of ME in 4 L of MR/d, n = 5) for 54 d after birth, at which point they were slaughtered and samples of mammary parenchyma tissue were obtained. Parenchymal mRNA was analyzed, and the fold change (FC) of 18,111 genes (ELE relative to RES) was uploaded to Ingenuity Pathway Analysis (IPA) software (Qiagen Bioinformatics, Redwood City, CA) for transcriptomic analysis. Using a threshold of P < 0.05, IPA identified that the FC of 1,931 of 18,811 differentially expressed genes (DEG) could be used for the analysis. A total of 18 molecular and cellular functions were relevant to DEG arising from the treatments; the 5 functions most associated with DEG were cell death and survival, cellular movement, cellular development, cellular growth and proliferation, and lipid metabolism. Based on the directional FC of DEG, the mammary gland of ELE heifers was predicted to have increased epithelial-mesenchymal transition (Z = 2.685) and accumulation of lipid (Z = 2.322), whereas the synthesis of DNA (Z = -2.137), transactivation of RNA (Z = -2.254), expression of RNA (Z = -2.405), transcription (Z = -2.482), and transactivation (Z = -2.611) were all predicted to be decreased. Additionally, IPA predicted the activation status of 13 upstream regulators with direct influence on DEG as affected by ELE feeding that were ligand-dependent nuclear receptors (n = 2), enzymes (n = 1), or transcription regulators (n = 10). Of these, 6 were activated (Z > 2) and 7 were inhibited (Z < -2). In summary, feeding ELE preweaning altered the mammary transcriptome of Holstein heifers, affecting cell functions involved in the morphological and physiological development of the mammary gland.
Collapse
Affiliation(s)
- K S Hare
- Department of Animal Bioscience, Animal Science and Nutrition, University of Guelph, Guelph, ON N1G 1Y2, Canada
| | - L N Leal
- R&D, Trouw Nutrition, PO Box 299, Amersfoort, 3800 AG, the Netherlands
| | - J M Romao
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Alberta, AB T6G 2P5, Canada
| | - G J Hooiveld
- Department of Agrotechnology and Food Sciences, Division of Human Nutrition and Health, Wageningen University, PO Box 17, Wageningen, 6700 AA, the Netherlands
| | - F Soberon
- Trouw Nutrition USA, Highland, IL 62249
| | - H Berends
- R&D, Trouw Nutrition, PO Box 299, Amersfoort, 3800 AG, the Netherlands
| | - M E Van Amburgh
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14850
| | - J Martín-Tereso
- R&D, Trouw Nutrition, PO Box 299, Amersfoort, 3800 AG, the Netherlands
| | - M A Steele
- Department of Animal Bioscience, Animal Science and Nutrition, University of Guelph, Guelph, ON N1G 1Y2, Canada.
| |
Collapse
|
7
|
Naderi S, Bohlouli M, Yin T, König S. Genomic breeding values, SNP effects and gene identification for disease traits in cow training sets. Anim Genet 2018; 49:178-192. [PMID: 29624705 DOI: 10.1111/age.12661] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2018] [Indexed: 12/30/2022]
Abstract
Holstein Friesian cow training sets were created according to disease incidences. The different datasets were used to investigate the impact of random forest (RF) and genomic BLUP (GBLUP) methodology on genomic prediction accuracies. In addition, for further verifications of some specific scenarios, single-step genomic BLUP was applied. Disease traits included the overall trait categories of (i) claw disorders, (ii) clinical mastitis and (iii) infertility from 80 741 first lactation Holstein cows kept in 58 large-scale herds. A subset of 6744 cows was genotyped (50K SNP panel). Response variables for all scenarios were de-regressed proofs (DRPs) and pre-corrected phenotypes (PCPs). Initially, all sick cows were allocated to the testing set, and healthy cows represented the training set. For the ongoing cow allocation schemes, the number of sick cows in the training set increased stepwise by moving 10% of the sick cows from the testing to the training set in each step. The size of training and testing sets was kept constant by replacing the same number of cows in the testing set with (randomly selected) healthy cows from the training set. For both the RF and GBLUP methods, prediction accuracies were larger for DRPs compared to PCPs. For PCPs as a response variable, the largest prediction accuracies were observed when the disease incidences in training sets reflected the disease incidence in the whole population. A further increase in prediction accuracies for some selected cow allocation schemes (i.e. larger prediction accuracies compared to corresponding scenarios with RF or GBLUB) was achieved via single-step GBLUP applications. Correlations between genome-wide association study SNP effects and RF importance criteria for single SNPs were in a moderate range, from 0.42 to 0.57, when considering SNPs from all chromosomes or from specific chromosome segments. RF identified significant SNPs close to potential positional candidate genes: GAS1, GPAT3 and CYP2R1 for clinical mastitis; SPINK5 and SLC26A2 for laminitis; and FGF12 for endometritis.
Collapse
Affiliation(s)
- S Naderi
- Institute of Animal Breeding and Genetics, University of Gießen, Ludwigstr. 21b, 35390, Gießen, Germany
| | - M Bohlouli
- Institute of Animal Breeding and Genetics, University of Gießen, Ludwigstr. 21b, 35390, Gießen, Germany
| | - T Yin
- Institute of Animal Breeding and Genetics, University of Gießen, Ludwigstr. 21b, 35390, Gießen, Germany
| | - S König
- Institute of Animal Breeding and Genetics, University of Gießen, Ludwigstr. 21b, 35390, Gießen, Germany
| |
Collapse
|
8
|
Akers RM. A 100-Year Review: Mammary development and lactation. J Dairy Sci 2017; 100:10332-10352. [DOI: 10.3168/jds.2017-12983] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/15/2017] [Indexed: 01/13/2023]
|
9
|
Rauner G, Barash I. Enrichment for Repopulating Cells and Identification of Differentiation Markers in the Bovine Mammary Gland. J Mammary Gland Biol Neoplasia 2016; 21:41-9. [PMID: 26615610 DOI: 10.1007/s10911-015-9348-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/15/2015] [Indexed: 10/22/2022] Open
Abstract
Elucidating cell hierarchy in the mammary gland is fundamental for understanding the mechanisms governing its normal development and malignant transformation. There is relatively little information on cell hierarchy in the bovine mammary gland, despite its agricultural potential and relevance to breast cancer research. Challenges in bovine-to-mouse xenotransplantation and difficulties obtaining bovine-compatible antibodies hinder the study of mammary stem-cell dynamics in this species. In-vitro indications of distinct bovine mammary epithelial cell populations, sorted according to CD24 and CD49f expression, have been provided. Here, we successfully transplanted these bovine populations into the cleared fat pads of immunocompromised mice, providing in-vivo evidence for the multipotency and self-renewal capabilities of cells that are at the top of the cell hierarchy (termed mammary repopulating units). Additional outgrowths from transplantation, composed exclusively of myoepithelial cells, were indicative of unipotent basal stem cells or committed progenitors. Sorting luminal cells according to E-cadherin revealed three distinct populations: luminal progenitors, and early- and late-differentiating cells. Finally, miR-200c expression was negatively correlated with differentiation levels in both the luminal and basal branches of the bovine mammary cell hierarchy. Together, these experiments provide further evidence for the presence of a regenerative entity in the bovine mammary gland and for the multistage differentiation process within the luminal lineage.
Collapse
Affiliation(s)
- Gat Rauner
- Institute of Animal Science, ARO, The Volcani Center, Bet-Dagan, Israel
- The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Itamar Barash
- Institute of Animal Science, ARO, The Volcani Center, Bet-Dagan, Israel.
| |
Collapse
|
10
|
Tucker HLM, Parsons CLM, Ellis S, Rhoads ML, Akers RM. Tamoxifen impairs prepubertal mammary development and alters expression of estrogen receptor α (ESR1) and progesterone receptors (PGR). Domest Anim Endocrinol 2016; 54:95-105. [PMID: 26619291 DOI: 10.1016/j.domaniend.2015.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/13/2015] [Accepted: 10/13/2015] [Indexed: 11/23/2022]
Abstract
Research has shown that prepubertal heifers experience allometric mammary growth that is influenced by the ovaries. Our purpose was to determine the role of estrogen in prepubertal mammary gland development. Sixteen Holstein calves were randomly assigned to 1 of 2 treatment groups: tamoxifen-injected (TAM) or control (CON). Calves were administered the antiestrogen tamoxifen (0.3 mg kg(1) d(1)) or placebo from 28 to 120 d of age. At 120 d, calves were euthanized and udders removed. Weight and DNA content of trimmed parenchymal tissue were halved (P ≤ 0.0001) in TAM compared with CON calves. Parenchymal samples from 3 zones of the left rear mammary gland (lower, middle, and outer regions) were processed for immunohistochemical staining for estrogen receptor α (ESR1) and progesterone receptor (PGR), Ki67-positive cells, and 5-bromo-2'-deoxyuridine label retaining cells (LRCs). Overall, neither the percentage nor location within the epithelial tissue layer of either ESR1- or PGR-positive cells was impacted by TAM treatment. However, image analysis indicated a 6.2-fold lower (P = 0.0001) level of ESR1 protein expression in TAM calves. Similarly, messenger RNA expression of ESR1 was also reduced (P = 0.0001) in TAM heifers. In contrast, expression of PGR protein was greater by 43% (P = 0.03) in TAM calves, but messenger RNA expression did not differ between treatments. Overall, TAM calves had a higher (P ≤ 0.03) percentage and density (cells per tissue area) of Ki67-positive cells. Irrespective of treatment, there were also more Ki67-labeled cells in the outer zones of the mammary gland (P ≤ 0.001). We were able to effectively use multispectral imaging to identify positive cells and quantify the expression of ESR1 and PGR protein. We also identified and counted the proportion of label retaining cells (LCR) (putative epithelial stem cells). We noted an overall 2.9-fold greater number of LRCs in TAM heifers and more LRCs in the outer sampling zones. This suggests that a cohort of LCR cells in TAM remained inactivated in comparison with CON heifers, which exhibited markedly increased growth of the mammary parenchymal tissue over the treatment period. These results suggest that the impacts of ovariectomy are partially explained by loss of ESR1 expression and/or estrogen receptor signaling in the prepubertal bovine mammary gland. The significance of mammary expression of PGR in control of prepubertal bovine mammary development remains unresolved.
Collapse
Affiliation(s)
- H L M Tucker
- Department of Dairy Science, Virginia Tech, Blacksburg, VA, 24061, USA
| | - C L M Parsons
- Department of Dairy Science, Virginia Tech, Blacksburg, VA, 24061, USA
| | - S Ellis
- Biological Sciences Directorate, National Science Foundation, Arlington, VA 22230, USA
| | - M L Rhoads
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - R M Akers
- Department of Dairy Science, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
11
|
Beaudry KL, Parsons CLM, Ellis SE, Akers RM. Localization and quantitation of macrophages, mast cells, and eosinophils in the developing bovine mammary gland. J Dairy Sci 2015; 99:796-804. [PMID: 26547646 DOI: 10.3168/jds.2015-9972] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/15/2015] [Indexed: 11/19/2022]
Abstract
Prepubertal mammary development involves elongation and branching of ducts and stromal tissue remodeling. This process is highly regulated and in mice is known to be affected by the presence of innate immune cells. Whether or not such immune cells are present or involved in bovine mammary development is unknown. For the first time, we determined the presence, location (relative to mammary ductal structures), and changes in numbers of eosinophils, mast cells, and macrophages in prepubertal bovine mammary tissue, and evaluated the effects of age, ovariectomy, and exogenous estrogen on numbers of each cell type. Chemical stains and immunofluorescence were used to identify the 3 cell types in formalin-fixed, paraffin-embedded mammary tissue from prepubertal female calves from 3 archived tissue sets. The ontogeny tissue set included samples of mammary tissue from female calves (n=4/wk) from birth to 6 wk of age. The ovary tissue set contained samples from ovary intact and ovariectomized heifers allowing us to investigate the influence of the ovaries on immune cells in the developing mammary gland in prepubertal heifers. Nineteen animals were intact or ovariectomized 30 d before sampling; they were 90, 120, or 150 d old at the time of sampling. A third tissue set, the estrogen set, allowed us to determine the effect of exogenous estrogen on innate immune cells in the gland. Eosinophils were identified via Luna staining, mast cells by May-Grunwald Giemsa staining, and macrophages with immunofluorescence. Key findings were that more eosinophils and mast cells were observed in near versus far stroma in the ontogeny and ovary tissue sets but not estrogen. More macrophages were observed in near versus far stroma in ontogeny animals. Eosinophils were more abundant in the younger animals, and fewer macrophages tended to be observed in ovariectomized heifers as compared with intact heifers and estrogen treatment resulted in a reduction in cell numbers. In summary, we show for the first time that innate immune cells are present in prepubertal bovine mammary tissue, localization varies by immune cell type, and abundance is related to proximity of epithelial structures and physiological state. We suggest a likely role for these cells in control of bovine mammary growth and ductal development.
Collapse
Affiliation(s)
- K L Beaudry
- Department of Dairy Sciences, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - C L M Parsons
- Department of Dairy Sciences, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - S E Ellis
- Department of Dairy Sciences, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - R M Akers
- Department of Dairy Sciences, Virginia Polytechnic Institute and State University, Blacksburg 24061.
| |
Collapse
|
12
|
Morphogenesis of Mammary Glands in Buffalo (Bubalus bubalis). ANATOMY RESEARCH INTERNATIONAL 2014; 2014:687936. [PMID: 24876967 PMCID: PMC4022206 DOI: 10.1155/2014/687936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 03/21/2014] [Accepted: 04/08/2014] [Indexed: 11/17/2022]
Abstract
The present research was elucidated on the morphogenesis of mammary gland of buffalo during prenatal development. Total of 16 foetuses ranging from 1.2 cm (34 days) to 108 cm CVRL (curved crown rump length) (317 days) were used for study. The study revealed that mammary line was first observed at 1.2 cm CVRL (34 days), mammary hillock at 1.7 cm (37 days), and mammary bud at 2.6 cm CVRL (41 days) foetuses. Epidermal cone was found at 6.7 cm CVRL (58 days) whereas primary and secondary ducts were observed at 7.4 cm CVRL (62 days) and 15 cm CVRL (96 days), respectively. Connective tissue whorls were reported at 18.2 cm CVRL (110 days) and internal elastic lamina and muscle layers at 24.1 cm CVRL (129 days). Lobules were observed at 29.3 cm CVRL (140 days), rosette of furstenberg at 39.5 cm CVRL (163 days), and keratin plug at 45.5 cm CVRL (176 days) foetus. Primordia of sweat and sebaceous glands around hair follicle were seen at 21.2 cm CVRL (122 days) of foetal life. Differentiation of all the skin layers along with cornification was observed at 69 cm (229 days) in group III foetuses.
Collapse
|
13
|
Comparison of stem/progenitor cell number and transcriptomic profile in the mammary tissue of dairy and beef breed heifers. J Appl Genet 2014; 55:383-95. [PMID: 24748329 PMCID: PMC4102771 DOI: 10.1007/s13353-014-0213-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 03/26/2014] [Accepted: 03/28/2014] [Indexed: 12/25/2022]
Abstract
Bovine mammary stem cells (MaSC) are a source of ductal and lobulo-alveolar tissue during the development of the mammary gland and its remodeling in repeating lactation cycles. We hypothesize that the number of MaSC, their molecular properties, and interactions with their niche may be essential in order to determine the mammogenic potential in heifers. To verify this hypothesis, we compared the number of MaSC and the transcriptomic profile in the mammary tissue of 20-month-old, non-pregnant dairy (Holstein-Friesian, HF) and beef (Limousin, LM) heifers. For the identification and quantification of putative stem/progenitor cells in mammary tissue sections, scanning cytometry was used with a combination of MaSC molecular markers: stem cell antigen-1 (Sca-1) and fibronectin type III domain containing 3B (FNDC3B) protein. Cytometric analysis revealed a significantly higher number of Sca-1posFNDC3Bpos cells in HF (2.94 ± 0.35 %) than in LM (1.72 ± 0.20 %) heifers. In HF heifers, a higher expression of intramammary hormones, growth factors, cytokines, chemokines, and transcription regulators was observed. The model of mammary microenvironment favorable for MaSC was associated with the regulation of genes involved in MaSC maintenance, self-renewal, proliferation, migration, differentiation, mammary tissue remodeling, angiogenesis, regulation of adipocyte differentiation, lipid metabolism, and steroid and insulin signaling. In conclusion, the mammogenic potential in postpubertal dairy heifers is facilitated by a higher number of MaSC and up-regulation of mammary auto- and paracrine factors representing the MaSC niche.
Collapse
|
14
|
Rauner G, Leviav A, Mavor E, Barash I. Development of Foreign Mammary Epithelial Morphology in the Stroma of Immunodeficient Mice. PLoS One 2013; 8:e68637. [PMID: 23825700 PMCID: PMC3688997 DOI: 10.1371/journal.pone.0068637] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 06/03/2013] [Indexed: 01/13/2023] Open
Abstract
Systemic growth and branching stimuli, and appropriate interactions with the host stroma are essential for the development of foreign epithelia in the mammary gland of immunodeficient mice. These factors were manipulated to promote and investigate the generation of representative bovine epithelial morphology in the transplanted mouse mammary stroma. The bovine mammary epithelium is unique in its commitment to rapid proliferation and high rate of differentiation. Its morphological organization within a fibrotic stroma resembles that of the human breast, and differs significantly from the rudimentary ductal network that penetrates a fatty stroma in mice. Transplantation of bovine mammary epithelial cells into the cleared mammary fat pad of NOD-SCID mice led to continuous growth of epithelial structures. Multilayered hollow spheres developed within fibrotic areas, but in contrast to mice, no epithelial organization was formed between adipocytes. The multilayered spheres shared characteristics with the heifer gland’s epithelium, including lumen size, cell proliferation, cytokeratin orientation, estrogen/progesterone receptor expression and localization, and milk protein synthesis. However, they did not extend into the mouse fat pad via ductal morphology. Pre-transplantation of fibroblasts increased the number of spheres, but did not promote extension of bovine morphology. The bovine cells preserved their fate and rarely participated in chimeric mouse–bovine outgrowths. Nevertheless, a single case of terminal ductal lobuloalveolar unit (TDLU) development was recorded in mice treated with estrogen and progesterone, implying the feasibility of this representative bovine morphology’s development. In vitro extension of these studies revealed paracrine inhibition of bovine epithelial mammosphere development by adipocytes, which was also generalized to breast epithelial mammosphere formation. The rescue of mammosphere development by fibroblast growth factor administration evidences an active equilibrium between inhibitory and supportive effects exerted by the adipose and fibrotic regions of the stroma, respectively, which determines the development of foreign epithelium.
Collapse
Affiliation(s)
- Gat Rauner
- Institute of Animal Science, ARO, The Volcani Center, Bet-Dagan, Israel
- The Robert H. Smith Faculty of Agriculture, Food and Environment, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amos Leviav
- Department of Plastic Surgery, Kaplan Medical Center, Rehovot, Israel
| | - Eliezer Mavor
- Department of Surgery, Kaplan Medical Center, Rehovot, Israel
| | - Itamar Barash
- Institute of Animal Science, ARO, The Volcani Center, Bet-Dagan, Israel
- * E-mail:
| |
Collapse
|
15
|
Borena BM, Bussche L, Burvenich C, Duchateau L, Van de Walle GR. Mammary stem cell research in veterinary science: an update. Stem Cells Dev 2013; 22:1743-51. [PMID: 23360296 DOI: 10.1089/scd.2012.0677] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The mammary gland is an organ with a remarkable regenerative capacity that can undergo multiple cycles of proliferation, lactation, and involution. Growing evidence suggests that these changes are driven by the coordinated division and differentiation of mammary stem cell populations (MaSC). Whereas information regarding MaSC and their role in comparative mammary gland physiology is readily available in human and mice, such information remains scarce in most veterinary mammal species such as cows, horses, sheep, goats, pigs, and dogs. We believe that a better knowledge on the MaSC in these species will not only help to gain more insights into mammary gland (patho) physiology in veterinary medicine, but will also be of value for human medicine. Therefore, this review summarizes the current knowledge on stem cell isolation and characterization in different mammals of veterinary importance.
Collapse
Affiliation(s)
- Bizunesh M Borena
- Department of Comparative Physiology and Biometrics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | | | | |
Collapse
|
16
|
Growth and development of the mammary glands of livestock: A veritable barnyard of opportunities. Semin Cell Dev Biol 2012; 23:557-66. [DOI: 10.1016/j.semcdb.2012.03.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 03/21/2012] [Accepted: 03/22/2012] [Indexed: 01/27/2023]
|
17
|
Cell hierarchy and lineage commitment in the bovine mammary gland. PLoS One 2012; 7:e30113. [PMID: 22253899 PMCID: PMC3258259 DOI: 10.1371/journal.pone.0030113] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 12/09/2011] [Indexed: 01/16/2023] Open
Abstract
The bovine mammary gland is a favorable organ for studying mammary cell hierarchy due to its robust milk-production capabilities that reflect the adaptation of its cell populations to extensive expansion and differentiation. It also shares basic characteristics with the human breast, and identification of its cell composition may broaden our understanding of the diversity in cell hierarchy among mammals. Here, Lin− epithelial cells were sorted according to expression of CD24 and CD49f into four populations: CD24medCD49fpos (putative stem cells, puStm), CD24negCD49fpos (Basal), CD24highCD49fneg (putative progenitors, puPgt) and CD24medCD49fneg (luminal, Lum). These populations maintained differential gene expression of lineage markers and markers of stem cells and luminal progenitors. Of note was the high expression of Stat5a in the puPgt cells, and of Notch1, Delta1, Jagged1 and Hey1 in the puStm and Basal populations. Cultured puStm and Basal cells formed lineage-restricted basal or luminal clones and after re-sorting, colonies that preserved a duct-like alignment of epithelial layers. In contrast, puPgt and Lum cells generated only luminal clones and unorganized colonies. Under non-adherent culture conditions, the puPgt and puStm populations generated significantly more floating colonies. The increase in cell number during culture provides a measure of propagation potential, which was highest for the puStm cells. Taken together, these analyses position puStm cells at the top of the cell hierarchy and denote the presence of both bi-potent and luminally restricted progenitors. In addition, a population of differentiated luminal cells was marked. Finally, combining ALDH activity with cell-surface marker analyses defined a small subpopulation that is potentially stem cell- enriched.
Collapse
|
18
|
Wang X, Reagan MR, Kaplan DL. Synthetic adipose tissue models for studying mammary gland development and breast tissue engineering. J Mammary Gland Biol Neoplasia 2010; 15:365-76. [PMID: 20835885 DOI: 10.1007/s10911-010-9192-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 08/24/2010] [Indexed: 12/20/2022] Open
Abstract
The mammary gland is a dynamic organ that continually changes its architecture and function. Reciprocal interactions between epithelium and adipocyte-containing stroma exert profound effects on all stages of its development, even though the details of these events are not fully understood. To address this issue, enormous potential exists in the utilization of synthetic adipose tissue model systems to uncover the properties and functions of adipocytes in the mammary gland. The first part of this review focuses on mammary adipose tissue (or adipocyte)-related model systems developed in recent years and their utility in investigating adipose-epithelial interactions, mammary gland morphogenesis, development and tumorigenesis. The second part shifts to the field of adipose-based breast tissue engineering, focusing on how these synthetic adipose tissue models are being constructed in vitro or in vivo for regeneration of the mammary gland, and their potentials in adipose tissue engineering also are discussed.
Collapse
Affiliation(s)
- Xiuli Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | | | | |
Collapse
|
19
|
Hovey RC, Aimo L. Diverse and active roles for adipocytes during mammary gland growth and function. J Mammary Gland Biol Neoplasia 2010; 15:279-90. [PMID: 20717712 PMCID: PMC2941079 DOI: 10.1007/s10911-010-9187-8] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 08/06/2010] [Indexed: 12/18/2022] Open
Abstract
The mammary gland is unique in its requirement to develop in close association with a depot of adipose tissue that is commonly referred to as the mammary fat pad. As discussed throughout this issue, the mammary fat pad represents a complex stromal microenvironment that includes a variety of cell types. In this article we focus on adipocytes as local regulators of epithelial cell growth and their function during lactation. Several important considerations arise from such a discussion. There is a clear and close interrelationship between different stromal tissue types within the mammary fat pad and its adipocytes. Furthermore, these relationships are both stage- and species-dependent, although many questions remain unanswered regarding their roles in these different states. Several lines of evidence also suggest that adipocytes within the mammary fat pad may function differently from those in other fat depots. Finally, past and future technologies present a variety of opportunities to model these complexities in order to more precisely delineate the many potential functions of adipocytes within the mammary glands. A thorough understanding of the role for this cell type in the mammary glands could present numerous opportunities to modify both breast cancer risk and lactation performance.
Collapse
Affiliation(s)
- Russell C Hovey
- Department of Animal Science, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA.
| | | |
Collapse
|
20
|
Punj V, Matta H, Chaudhary PM. X-linked ectodermal dysplasia receptor is downregulated in breast cancer via promoter methylation. Clin Cancer Res 2010; 16:1140-8. [PMID: 20145163 PMCID: PMC2822888 DOI: 10.1158/1078-0432.ccr-09-2463] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The X-linked ectodermal dysplasia receptor (XEDAR) is a novel receptor of the tumor necrosis factor receptor family that binds to ectodysplasin-A2 (EDA-A2) and induces cell death. The purpose of this study was to determine the tumor-suppressive potential of XEDAR in the development of breast cancer. EXPERIMENTAL DESIGN We analyzed the expression of XEDAR in breast cancer cell lines and tumor samples using quantitative real-time PCR analysis and immunoblotting. We analyzed the human XEDAR gene promoter for the presence of any CpG island and examined its methylation status using methylation-specific real-time PCR. We examined the effect of 5-aza-2'-deoxycytidine on the expression of XEDAR and sensitivity to EDA-A2-induced apoptosis in breast cancer cell lines. RESULTS Expression of XEDAR, but not EDA-A2, was downregulated in most tumorigenic breast cancer cell lines and tumor samples. Loss of XEDAR expression correlated with the hypermethylation of its promoter. Ectopic expression of XEDAR in MDA-MB-231 cells resulted in significant induction of apoptosis and reduction in colony formation. Treatment with 5-aza-2'-deoxycytidine restored XEDAR expression in breast cancer cell lines with methylated XEDAR promoter and sensitized them to EDA-A2-induced cell death. CONCLUSIONS Our results suggest that XEDAR expression is downregulated in most breast cancers via promoter methylation, which may contribute to accelerated tumor development by blocking EDA-A2-induced cell death. XEDAR may represent a novel breast tumor suppressor gene, and restoration of its expression by treatment with DNA demethylating agents may represent an attractive approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Vasu Punj
- Department of Medicine, Division of Hematology-Oncology, Hillman Cancer Center, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15213-1863, USA.
| | | | | |
Collapse
|
21
|
Marshall AM, Pai VP, Sartor MA, Horseman ND. In vitro multipotent differentiation and barrier function of a human mammary epithelium. Cell Tissue Res 2008; 335:383-95. [PMID: 19005683 DOI: 10.1007/s00441-008-0719-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Accepted: 10/06/2008] [Indexed: 12/16/2022]
Abstract
As demonstrated by a variety of animal studies, barrier function in the mammary epithelium is essential for a fully functioning and differentiated gland. However, there is a paucity of information on barrier function in human mammary epithelium. Here, we have established characteristics of a polarizing differentiating model of human mammary epithelial cells capable of forming a high-resistance/low-conductance barrier in a predictable manner, viz., by using MCF10A cells on permeable membranes. Inulin flux decreased and transepithelial electrical resistance (TEER) increased over the course of several days after seeding MCF10A cells on permeable membranes. MCF10A cells exhibited multipotent phenotypic differentiation into layers expressing basal and lumenal markers when placed on permeable membranes, with at least two distinct cell phenotypes. A clonal subline of MCF10A, generated by culturing stem-like cells under non-adherent conditions, also generated a barrier-forming epithelial membrane with cells expressing markers of both basal and lumenal differentiation (CD10 and MUC1, respectively). Progressive changes associated with differentiation, including wholesale inhibition of cell-cycle genes and stimulation of cell and tissue morphogenic genes, were observed by gene expression profiling. Clustering and gene ontology categorization of significantly altered genes revealed a pattern of lumenal epithelial-cell-specific differentiation.
Collapse
Affiliation(s)
- Aaron M Marshall
- Systems Biology and Physiology Program, Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH 45267-0576, USA
| | | | | | | |
Collapse
|
22
|
Lewis CM, Herbert BS, Bu D, Halloway S, Beck A, Shadeo A, Zhang C, Ashfaq R, Shay JW, Euhus DM. Telomerase immortalization of human mammary epithelial cells derived from a BRCA2 mutation carrier. Breast Cancer Res Treat 2006; 99:103-15. [PMID: 16541310 DOI: 10.1007/s10549-006-9189-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2005] [Accepted: 02/05/2006] [Indexed: 12/13/2022]
Abstract
A novel human mammary epithelial cell line, HME348, was established from benign breast tissue from a 44-year-old germ-line BRCA2 mutation carrier with a history of stage 1 breast cancer. Mutation analysis showed that the patient had a known 6872del4 BRCA2 heterozygous mutation. The human mammary epithelial cells passaged in culture exhibited cellular replicative aging as evidenced by telomere shortening, lack of telomerase activity, and senescence. Ectopic expression of telomerase (hTERT) reconstituted telomerase activity in these cells and led to the immortalization of the cells. When grown on glass, the majority of immortalized HME348 cells expressed ESA and p63 with a small population also expressing EMA. In three-dimensional Matrigel culture, HME348 cells formed complex branching acini structures that expressed luminal (EMA, CK18) and myoepithelial (p63, CALLA, CK14) markers. Three clones derived from this culture were also p63(+)/ESA(+)/EMA(+/-) on glass but formed similar acinar structures with both luminal and myoepithelial cell differentiation in Matrigel confirming the mammary progenitor nature of these cells. Additionally, the experimentally immortalized HME348 cells formed acini in cleared mammary fat pads in vivo. As this is the first report establishing and characterizing a benign human mammary epithelial cell line derived from a BRCA2 patient without the use of viral oncogenes, these cells may be useful for the study of BRCA2 function in breast morphogenesis and carcinogenesis.
Collapse
Affiliation(s)
- Cheryl M Lewis
- Hamon Center for Therapeutic Oncology Research and Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390-9039, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Three-dimensional (3D) tissue culture models have an invaluable role in tumour biology today providing some very important insights into cancer biology. As well as increasing our understanding of homeostasis, cellular differentiation and tissue organization they provide a well defined environment for cancer research in contrast to the complex host environment of an in vivo model. Due to their enormous potential 3D tumour cultures are currently being exploited by many branches of biomedical science with therapeutically orientated studies becoming the major focus of research. Recent advances in 3D culture and tissue engineering techniques have enabled the development of more complex heterologous 3D tumour models.
Collapse
Affiliation(s)
- Jong Bin Kim
- Ludwig Institute for Cancer Research, First Floor - Breast Cancer Laboratory, Department of Surgery, Royal Free and University College London Medical School, Charles Bell House, 67-73 Riding House Street, London W1W 7EJ, UK.
| |
Collapse
|
24
|
Vangroenweghe F, Lamote I, Burvenich C. Physiology of the periparturient period and its relation to severity of clinical mastitis. Domest Anim Endocrinol 2005; 29:283-93. [PMID: 15950428 DOI: 10.1016/j.domaniend.2005.02.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Revised: 02/16/2005] [Accepted: 02/19/2005] [Indexed: 10/25/2022]
Abstract
Incidence of clinical mastitis is highest at drying off and during the periparturient period. Intramammary Escherichia coli infection in high-yielding cows can show a severe clinical response during the early post-partum period. Severe clinical mastitis is mainly determined by cow factors, in particular the functionality of the circulating polymorphonuclear leukocytes (PMN) which are recruited to the mammary gland during the inflammatory reaction. There is a co-incidence between the periods of highest incidence of clinical mastitis and specific structural changes in the mammary gland. During the periparturient period, marked changes in various systemic and local hormones are related to the secretory state of the mammary gland epithelium (lactogenesis). Estrogen and progesterone induce proliferation of the mammary epithelium throughout gestation and act as survival factors in different tissues, although conflicting data have been reported on their effect on PMN oxidative burst. Somatotropin (STH), responsible for maintenance of lactation in ruminants, has been shown to positively influence innate immunity and a more rapid recovery in milk production of severely affected animals. The concentration of STH, and as a result also IGF-I levels is, however, quite low during early lactation. IGF-I and its regulating binding proteins are associated with cell survival, modulation of apoptosis and functionality of PMN in humans. During early lactation, bio-availability of IGF-I is decreased, which might reduce its stimulating effects on PMN quality and functionality. PRL, concomitantly known as a lactogenic hormone and an immunoregulatory cytokine, has also been associated with modulation of the immune system. It is expected that in periparturient animals, hormone changes could interfere with the immune response and the clinical response of mastitis.
Collapse
Affiliation(s)
- F Vangroenweghe
- Milk Secretion and Mastitis Research Center, Department of Physiology-Biochemistry-Biometrics, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | | | | |
Collapse
|
25
|
Hariri M, Wood GA, DiGrappa MA, MacPherson M, Backman SA, Yaffe MJ, Mak TW, Boyd NF, Khokha R. Experimental manipulation of radiographic density in mouse mammary gland. Breast Cancer Res 2004; 6:R540-5. [PMID: 15318935 PMCID: PMC549169 DOI: 10.1186/bcr901] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Revised: 05/12/2004] [Accepted: 06/01/2004] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Extensive mammographic density in women is associated with increased risk for breast cancer. Mouse models provide a powerful approach to the study of human diseases, but there is currently no model that is suited to the study of mammographic density. METHODS We performed individual manipulations of the stromal, epithelial and matrix components of the mouse mammary gland and examined the alterations using in vivo and ex vivo radiology, whole mount staining and histology. RESULTS Areas of density were generated that resembled densities in mammographic images of the human breast, and the nature of the imposed changes was confirmed at the cellular level. Furthermore, two genetic models, one deficient in epithelial structure (Pten conditional tissue specific knockout) and one with hyperplastic epithelium and mammary tumors (MMTV-PyMT), were used to examine radiographic density. CONCLUSION Our data show the feasibility of altering and imaging mouse mammary gland radiographic density by experimental and genetic means, providing the first step toward modelling the biological processes that are responsible for mammographic density in the mouse.
Collapse
Affiliation(s)
- Mehrdad Hariri
- Department of Medical Biophysics, Ontario Cancer Institute/University Health Network, Toronto, Ontario, Canada
| | - Geoffrey A Wood
- Department of Medical Biophysics, Ontario Cancer Institute/University Health Network, Toronto, Ontario, Canada
| | - Marco A DiGrappa
- Department of Medical Biophysics, Ontario Cancer Institute/University Health Network, Toronto, Ontario, Canada
| | - Michelle MacPherson
- Imaging Research, Sunnybrook and Women's Health Sciences Centre, Toronto, Ontario, Canada
| | - Stephanie A Backman
- Department of Medical Biophysics, Ontario Cancer Institute/University Health Network, Toronto, Ontario, Canada
| | - Martin J Yaffe
- Imaging Research, Sunnybrook and Women's Health Sciences Centre, Toronto, Ontario, Canada
| | - Tak W Mak
- Department of Medical Biophysics, Ontario Cancer Institute/University Health Network, Toronto, Ontario, Canada
| | - Norman F Boyd
- Department of Medical Biophysics, Ontario Cancer Institute/University Health Network, Toronto, Ontario, Canada
| | - Rama Khokha
- Department of Medical Biophysics, Ontario Cancer Institute/University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Kim JB, Stein R, O'Hare MJ. Three-dimensional in vitro tissue culture models of breast cancer-- a review. Breast Cancer Res Treat 2004; 85:281-91. [PMID: 15111767 DOI: 10.1023/b:brea.0000025418.88785.2b] [Citation(s) in RCA: 217] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Three-dimensional (3D) in vitro breast tumour models have an invaluable role in tumour biology today providing some very important insights into breast cancer. As well as increasing our understanding of homeostasis, cellular differentiation and tissue organization they provide a well defined environment for cancer research in contrast to the complex host environment of an in vivo model. With the recent availability of relevant stromal elements together with the vast array of extracellular matrix constituents available, in vivo like microenvironments can be recreated. These tissue like structures more realistically model the structural architecture and differentiated function of breast cancer than a cellular monolayer providing in vivo like responses to therapeutic agents. Three dimensional in vitro models allow the study of cell-cell and cell-extracellular matrix interactions, in addition to the influence of the microenvironment on cellular differentiation, proliferation, apoptosis and gene expression. Due to their enormous potential 3D cultures are currently being exploited by many other branches of biomedical science with therapeutically orientated studies becoming the major focus of research. In return great progress in 3D culture techniques have been made, largely due to this greater interaction. At present they are being used in studies ranging from investigating the role of adhesion molecules (e.g., E-cadherin) in invasion/metastasis; VEGF and angiogenesis, to tissue modelling and remodelling. Progress in the development of complex 3D culture systems is more productive than ever, however further research is vital.
Collapse
Affiliation(s)
- Jong Bin Kim
- Department of Surgery, Royal Free and University College London Medical School, Ludwig Institute for Cancer Research/University College London, Charles Bell House, London, UK.
| | | | | |
Collapse
|
27
|
Schmeichel KL, Bissell MJ. Modeling tissue-specific signaling and organ function in three dimensions. J Cell Sci 2003; 116:2377-88. [PMID: 12766184 PMCID: PMC2933213 DOI: 10.1242/jcs.00503] [Citation(s) in RCA: 416] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In order to translate the findings from basic cellular research into clinical applications, cell-based models need to recapitulate both the 3D organization and multicellular complexity of an organ but at the same time accommodate systematic experimental intervention. Here we describe a hierarchy of tractable 3D models that range in complexity from organotypic 3D cultures (both monotypic and multicellular) to animal-based recombinations in vivo. Implementation of these physiologically relevant models, illustrated here in the context of human epithelial tissues, has enabled the study of intrinsic cell regulation pathways and also has provided compelling evidence for the role of the stromal compartment in directing epithelial cell function and dysfunction. Furthermore the experimental accessibility afforded by these tissue-specific 3D models has implications for the design and development of cancer therapies.
Collapse
Affiliation(s)
- Karen L Schmeichel
- Lawrence Berkeley National Laboratory, 1 Cyclotron Road, MS 83-101, CA 94720, USA.
| | | |
Collapse
|
28
|
Hovey RC, Trott JF, Vonderhaar BK. Establishing a framework for the functional mammary gland: from endocrinology to morphology. J Mammary Gland Biol Neoplasia 2002; 7:17-38. [PMID: 12160083 DOI: 10.1023/a:1015766322258] [Citation(s) in RCA: 218] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
From its embryonic origins, the mammary gland in females undergoes a course of ductal development that supports the establishment of alveolar structures during pregnancy prior to the onset of lactogenesis. This development includes multiple stages of proliferation and morphogenesis that are largely directed by concurrent alterations in key hormones and growth factors across various reproductive states. Ductal elongation is directed by estrogen, growth hormone, insulin-like growth factor-I, and epidermal growth factor, whereas ductal branching and alveolar budding is influenced by additional factors such as progesterone, prolactin, and thyroid hormone. The response by the ductal epithelium to various hormones and growth factors is influenced by epithelial-stromal interactions that differ between species, possibly directing species-specific morphogenesis. Evolving technologies continue to provide the opportunity to further delineate the regulation of ductal development. Defining the hormonal control of ductal development should facilitate a better understanding of the mechanisms underlying mammary gland tumorigenesis.
Collapse
Affiliation(s)
- Russell C Hovey
- Molecular and Cellular Endocrinology Section, Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1402, USA
| | | | | |
Collapse
|
29
|
Hovey RC, Davey HW, Vonderhaar BK, Mackenzie DD, McFadden TB. Paracrine action of keratinocyte growth factor (KGF) during ruminant mammogenesis. Mol Cell Endocrinol 2001; 181:47-56. [PMID: 11476940 DOI: 10.1016/s0303-7207(01)00537-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Keratinocyte growth factor (KGF) is a stroma-derived mitogen mediating epithelial-stromal interactions. We investigated the role of KGF during epithelial-stromal interactions accompanying ruminant mammogenesis. Target-specificity of KGF was demonstrated in that KGF-stimulated proliferation of bovine mammary epithelial, but not ovine mammary stromal cells. Consistent with a paracrine function, 4.6, 2.4, 1.5 and 0.9 kb mRNA transcripts were expressed by bovine stromal, but not epithelial cells. Within the ovine mammary gland, 2.4 and 1.5 kb KGF mRNAs were expressed in the fat pad while only the 2.4 kb transcript was transcribed in parenchyma. The abundance of KGF mRNA was greater in the extra-parenchymal fat pad than in the contralateral epithelium-free fat pad prior to puberty, and was less in parenchyma than in the intact or epithelium-free fat pads. Ovariectomy tended to increase KGF transcription while estrogen reduced expression. Of several tissues, mammary parenchyma expressed a 2.4 kb mRNA while adipose tissues expressed a 1.5 kb transcript. These results demonstrate local and systemic regulation of KGF transcription and support a paracrine role for KGF during ruminant mammogenesis.
Collapse
Affiliation(s)
- R C Hovey
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
30
|
Akers RM, McFadden TB, Purup S, Vestergaard M, Sejrsen K, Capuco AV. Local IGF-I axis in peripubertal ruminant mammary development. J Mammary Gland Biol Neoplasia 2000; 5:43-51. [PMID: 10791767 DOI: 10.1023/a:1009563115612] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The regulation of mammary growth and development in heifers is accomplished by complex interactions of hormones, growth factors, and extracellular matrix molecules. Many of these growth stimulators are believed to be locally produced in the mammary gland and to be affected by developmental and nutritional status. Although estrogen and growth hormone are considered critical to pubertal mammogenesis, results summarized in this review suggest that IGF-I and IGF binding proteins are especially important locally-produced growth regulators in peripubertal ruminants. This assertion is supported by studies of ovariectomized heifers, in which increased stromal IGFBP-3 and reduced IGF-I correspond with a failure of udder development. Similarly, reduced mammary development with overfeeding coincides with reduced mitogenic activity of mammary tissue extracts and altered concentrations of IGF-I and IGFBPs. In vitro studies convincingly demonstrate that much of the mitogenic activity of mammary extracts or serum can be attributed to IGF-I and that alterations in IGFBP-3 modulate its effectiveness. Thus by analogy to second messenger mechanisms of action for protein hormones, local mammary-derived growth factors likely explain many of the effects attributed to the classic mammogenic hormones.
Collapse
Affiliation(s)
- R M Akers
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Hovey RC, McFadden TB, Akers RM. Regulation of mammary gland growth and morphogenesis by the mammary fat pad: a species comparison. J Mammary Gland Biol Neoplasia 1999; 4:53-68. [PMID: 10219906 DOI: 10.1023/a:1018704603426] [Citation(s) in RCA: 158] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The growth and morphogenesis of mammary parenchyma varies substantially between species and is regulated by an array of systemic and local factors. Central to this regulation is the mammary fat pad, a matrix of adipose and connective tissue capable of mediating hormone action and synthesizing an array of growth regulatory molecules. In this article we highlight differences between the morphological development of the mammary parenchyma in rodents, humans, and ruminant dairy animals, placing emphasis on differences in the cellular composition and structure of the mammary fat pad. While a great deal remains to be understood about the ability of stroma to locally regulate mammary development, the significance of its contribution is becoming increasingly apparent. The actions of several steroid and peptide hormones appear to be mediated by an array of growth factors, proteases and extracellular matrix components synthesized by constituents of the mammary fat pad. Further, mammary adipose tissue represents a significant store of lipid which, by itself and through its derivatives, could influence the growth of mammary epithelium in diverse ways. This review describes the integral role of the mammary fat pad during mammogenesis, emphasizing the point that species differences must be addressed if local growth and morphogenic mechanisms within the mammary gland are to be resolved.
Collapse
Affiliation(s)
- R C Hovey
- Laboratory of Tumor Immunology and Biology, National Institutes of Health, Bethesda, Maryland 20892-1402, USA.
| | | | | |
Collapse
|
32
|
Abstract
The mammary fat pad is essential for development of the mammary epithelium, providing signals that mediate ductal morphogenesis and, probably, alveolar differentiation. The "cleared" fat pad is often used as a transplantation site. Considering the crucial role of the fat pad, its properties have received relatively little attention from researchers in the field. Some of the questions whose investigation is pertinent to understanding both normal mammary development and carcinogenesis are outlined in this commentary in the spirit of stimulating enquiry into this important subject. It is clear from a brief perusal of the available literature that until studies are specifically designed to clearly differentiate between functional effects of the fibrous and the adipose stroma, more substantive information about their differential effects on mammary development and tumorigenesis will not be forthcoming.
Collapse
|
33
|
Abstract
The ability to store substantial amounts of energy as lipid in adipose tissue has allowed development of a variety of strategies in wild animals to meet the considerable metabolic challenge of lactation. The ability to use adipose tissue energy has also been critical for development of the exceptional rates of milk production achieved in the dairy cow. Lactation thus results in profound changes in adipose tissue metabolism, the molecular bases of which are beginning to be resolved in domestic ruminants and laboratory rodents. In addition to its role as an energy store, adipose tissue has a variety of other functions (e.g., modulation of mammary development, appetite, immune system function), some of which are important for lactation.
Collapse
Affiliation(s)
- R G Vernon
- Hannah Research Institute, Ayr, Scotland, United Kingdom
| | | |
Collapse
|
34
|
Nguyen B, Keane MM, Johnston PG. The biology of growth regulation in normal and malignant breast epithelium: from bench to clinic. Crit Rev Oncol Hematol 1995; 20:223-36. [PMID: 8748011 DOI: 10.1016/1040-8428(94)00161-l] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- B Nguyen
- NCI-Navy Medical Oncology Branch, Division of Cancer Treatment, National Cancer Institute, Bethesda, MD, USA
| | | | | |
Collapse
|
35
|
Clarke R, Dickson RB, Lippman ME. Hormonal aspects of breast cancer. Growth factors, drugs and stromal interactions. Crit Rev Oncol Hematol 1992; 12:1-23. [PMID: 1540336 DOI: 10.1016/1040-8428(92)90062-u] [Citation(s) in RCA: 107] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- R Clarke
- Lombardi Cancer Research Center, Georgetown University Medical Center, Washington DC 20007
| | | | | |
Collapse
|