Sequencing the transcriptome of milk production: milk trumps mammary tissue

Danielle G Lemay, Russell C Hovey, Stella R Hartono, Katie Hinde, Jennifer T Smilowitz, Frank Ventimiglia, Kimberli A Schmidt, Joyce W S Lee, Alma Islas-Trejo, Pedro Ivo Silva, Ian Korf, Juan F Medrano, Peter A Barry, J Bruce German, Danielle G Lemay, Russell C Hovey, Stella R Hartono, Katie Hinde, Jennifer T Smilowitz, Frank Ventimiglia, Kimberli A Schmidt, Joyce W S Lee, Alma Islas-Trejo, Pedro Ivo Silva, Ian Korf, Juan F Medrano, Peter A Barry, J Bruce German

Abstract

Background: Studies of normal human mammary gland development and function have mostly relied on cell culture, limited surgical specimens, and rodent models. Although RNA extracted from human milk has been used to assay the mammary transcriptome non-invasively, this assay has not been adequately validated in primates. Thus, the objectives of the current study were to assess the suitability of lactating rhesus macaques as a model for lactating humans and to determine whether RNA extracted from milk fractions is representative of RNA extracted from mammary tissue for the purpose of studying the transcriptome of milk-producing cells.

Results: We confirmed that macaque milk contains cytoplasmic crescents and that ample high-quality RNA can be obtained for sequencing. Using RNA sequencing, RNA extracted from macaque milk fat and milk cell fractions more accurately represented RNA from mammary epithelial cells (cells that produce milk) than did RNA from whole mammary tissue. Mammary epithelium-specific transcripts were more abundant in macaque milk fat, whereas adipose or stroma-specific transcripts were more abundant in mammary tissue. Functional analyses confirmed the validity of milk as a source of RNA from milk-producing mammary epithelial cells.

Conclusions: RNA extracted from the milk fat during lactation accurately portrayed the RNA profile of milk-producing mammary epithelial cells in a non-human primate. However, this sample type clearly requires protocols that minimize RNA degradation. Overall, we validated the use of RNA extracted from human and macaque milk and provided evidence to support the use of lactating macaques as a model for human lactation.

Figures

Figure 1
Figure 1
Composite images of whole milk stained with acridine orange (AO). Each image contains three channels: 1) a differential interference contrast image to view the fat globules, which look like gray bubbles; 2) a fluorescence channel for AO-RNA; and 3) another fluorescence channel for AO-DNA. When AO associates with RNA, the emission maximum is 650 nm (red). When AO associates with DNA, the emission maximum is 525 nm (green). Therefore, areas containing only RNA (e.g. crescents) look red, only DNA look green, and both RNA and DNA (e.g. nucleated cells) look yellow. (A) Close-up of human whole milk with a nucleated cell (yellow) and several crescents of various sizes. (B) Whole slide scanned image of human whole milk collected after a 4-hr milk accumulation. (C–D) Whole slide scanned images of macaque whole milk after (C) a 4-hr and (D) a 10-min milk accumulation.
Figure 2
Figure 2
Percentage of cytoplasmic crescents and cells in milk. Milk samples were collected after a 4-hr accumulation period in human subjects and a 4-hr, 2-hr, or 10-min accumulation in macaques. The boxplots show the (A) percentage of milk fat globules with cytoplasmic crescents and (B) the percentage of RNA attributable to nucleated cells in these milk samples. (A-B) The upper and lower bounds of each box denote the first and third quartiles of the data, the dark horizontal line within each box denotes the median value, and the whiskers extend to the minimum and maximum values. The width of each box is proportional to the square root of the samples size. Asterisks denote statistical significance (p < = 0.05).
Figure 3
Figure 3
Effect of milk fraction and processing time on RNA integrity. (A-B) RNA degradation by sample types in (A) human and (B) macaque samples. All differences are significant (t-test, p < = 0.05). (C-D) Human milk fat RNA degradation by processing time in terms of (C) the distance to the lab from the site of collection (R2 = 0.40, p = 0.012) and (D) the amount of storage time in TRIzol at -80°C prior to RNA isolation (R2 = 0.26, p = 0.044).
Figure 4
Figure 4
Hematoxylin- and eosin-stained mammary biopsy samples from rhesus macaques. Histology of tissue biopsied from the mammary glands of rhesus macaques at different stages of lactation. Sections (4 μm) were stained with hematoxylin and eosin. Scale bar = 100 μm. (A) Multiparous aged female macaque, non-pregnant, non-lactating; (B) multiparous female macaque, lactation day 30; and (C) multiparous female macaque, lactation day 90.
Figure 5
Figure 5
Expression of cell-specific markers in milk and mammary transcriptomes. Each box in the heatmap represents log-transformed expression intensity. The color key indicates the level of expression: Green = low/no expression, Yellow = low expression, Orange = moderate expression, Pink = high expression, and White = very high expression. The color key also includes a light blue line that indicates the number of observations at each level of expression.
Figure 6
Figure 6
Abundances of transcripts that code for proteins in the lactose synthesis pathway. All differences between mammary biopsy (“Bx”) and milk fat samples (“Fat”) were significant. Differences between milk fat (“Fat”) and milk cell (“Cells”) samples were significant for all transcripts except LALBA and B4GALT.
Figure 7
Figure 7
Transcriptome homogeneity within sample types. Transcriptomes derived from samples of the same type (mammary biopsy, milk cells, or milk fat) were compared with other samples of the same type using a Spearman’s correlation. The boxplot summarizes the findings by sample type. The upper and lower bounds of each box denote the first and third quartiles of the data, the dark horizontal line within each box denotes the median value, and the whiskers extend to the minimum and maximum values. The width of each box is proportional to the square root of the samples size. Asterisks denote statistical significance (p < = 0.05).
Figure 8
Figure 8
Correlation of milk and mammary transcriptomes. Each square in the heatmap represents the pairwise correlation of the sample listed in the row compared with the sample listed in the column. The shades of blue across the top of each heatmap correspond to sample type. The colors on the left side of each heat map correspond to each individual monkey. Both biological and technical replicates are shown. All technical replicates appear as clustered triplets in the dendograms. (A) Milk fat vs mammary biopsy samples; dark blue denotes biopsy, light blue denotes milk fat. (B) Milk cells vs mammary biopsy samples, dark blue denotes milk cell, light blue denotes milk fat. (C) Milk cells vs milk fat samples, dark blue denotes milk cell, blue denotes biopsy. (D) Mammary biopsy samples at day 30 and day 90. (E) Milk fat samples at day 30 and day 90. (F) Milk cells samples at day 30 and day 90. (D–F) Dark and light blue distinguish day 30 and 90 samples.

References

    1. Rudolph MC, McManaman JL, Hunter L, Phang T, Neville MC. Functional development of the mammary gland: use of expression profiling and trajectory clustering to reveal changes in gene expression during pregnancy, lactation, and involution. J Mammary Gland Biol Neoplasia. 2003;8(3):287–307.
    1. Stein T, Morris JS, Davies CR, Weber-Hall SJ, Duffy MA, Heath VJ, Bell AK, Ferrier RK, Sandilands GP, Gusterson BA. Involution of the mouse mammary gland is associated with an immune cascade and an acute-phase response, involving LBP, CD14 and STAT3. Breast cancer research: BCR. 2004;6(2):R75–R91. doi: 10.1186/bcr753.
    1. Ron M, Israeli G, Seroussi E, Weller JI, Gregg JP, Shani M, Medrano JF. Combining mouse mammary gland gene expression and comparative mapping for the identification of candidate genes for QTL of milk production traits in cattle. BMC Genomics. 2007;8:183. doi: 10.1186/1471-2164-8-183.
    1. Ramanathan P, Martin IC, Gardiner-Garden M, Thomson PC, Taylor RM, Ormandy CJ, Moran C, Williamson P. Transcriptome analysis identifies pathways associated with enhanced maternal performance in QSi5 mice. BMC Genomics. 2008;9:197. doi: 10.1186/1471-2164-9-197.
    1. Wei J, Ramanathan P, Martin IC, Moran C, Taylor RM, Williamson P. Identification of gene sets and pathways associated with lactation performance in mice. Physiol Genomics. 2013;45(5):171–181. doi: 10.1152/physiolgenomics.00139.2011.
    1. Suchyta SP, Sipkovsky S, Halgren RG, Kruska R, Elftman M, Weber-Nielsen M, Vandehaar MJ, Xiao L, Tempelman RJ, Coussens PM. Bovine mammary gene expression profiling using a cDNA microarray enhanced for mammary-specific transcripts. Physiol Genomics. 2003;16(1):8–18. doi: 10.1152/physiolgenomics.00028.2003.
    1. Finucane KA, McFadden TB, Bond JP, Kennelly JJ, Zhao FQ. Onset of lactation in the bovine mammary gland: gene expression profiling indicates a strong inhibition of gene expression in cell proliferation. Functional & integrative genomics. 2008;8(3):251–264. doi: 10.1007/s10142-008-0074-y.
    1. Rudolph MC, McManaman JL, Phang T, Russell T, Kominsky DJ, Serkova NJ, Stein T, Anderson SM, Neville MC. Metabolic regulation in the lactating mammary gland: a lipid synthesizing machine. Physiol Genomics. 2007;28(3):323–336.
    1. Anderson SM, Rudolph MC, McManaman JL, Neville MC. Key stages in mammary gland development. Secretory activation in the mammary gland: it’s not just about milk protein synthesis! Breast Cancer Res: BCR. 2007;9(1):204. doi: 10.1186/bcr1653.
    1. Wall EH, Bond JP, McFadden TB. Acute milk yield response to frequent milking during early lactation is mediated by genes transiently regulated by milk removal. Physiol Genomics. 2012;44(1):25–34. doi: 10.1152/physiolgenomics.00027.2011.
    1. Brand B, Hartmann A, Repsilber D, Griesbeck-Zilch B, Wellnitz O, Kuhn C, Ponsuksili S, Meyer HH, Schwerin M. Comparative expression profiling of E. coli and S. aureus inoculated primary mammary gland cells sampled from cows with different genetic predispositions for somatic cell score. Genetics, selection, evolution : GSE. 2011;43:24. doi: 10.1186/1297-9686-43-24.
    1. Wall EH, Bond JP, McFadden TB. Milk yield responses to changes in milking frequency during early lactation are associated with coordinated and persistent changes in mammary gene expression. BMC Genomics. 2013;14(1):296. doi: 10.1186/1471-2164-14-296.
    1. Bionaz M, Periasamy K, Rodriguez-Zas SL, Everts RE, Lewin HA, Hurley WL, Loor JJ. Old and new stories: revelations from functional analysis of the bovine mammary transcriptome during the lactation cycle. PLoS ONE. 2012;7(3):e33268. doi: 10.1371/journal.pone.0033268.
    1. Moyes KM, Drackley JK, Morin DE, Rodriguez-Zas SL, Everts RE, Lewin HA, Loor JJ. Mammary gene expression profiles during an intramammary challenge reveal potential mechanisms linking negative energy balance with impaired immune response. Physiol Genomics. 2010;41(2):161–170. doi: 10.1152/physiolgenomics.00197.2009.
    1. Medrano JF, Rincon G, Islas-Trejo A. 9th World Congress on Genetics applied to Livestock Production: 2010. Leipzig, Germany; 2010. Comparative analysis of bovine milk and mammary gland transcriptome using RNA-Seq; p. 852. .
    1. Wickramasinghe S, Hua S, Rincon G, Islas-Trejo A, German JB, Lebrilla CB, Medrano JF. Transcriptome profiling of bovine milk oligosaccharide metabolism genes using RNA-sequencing. PLoS ONE. 2011;6(4):e18895. doi: 10.1371/journal.pone.0018895.
    1. Wickramasinghe S, Rincon G, Islas-Trejo A, Medrano JF. Transcriptional profiling of bovine milk using RNA sequencing. BMC Genomics. 2012;13:45. doi: 10.1186/1471-2164-13-45.
    1. Canovas A, Rincon G, Islas-Trejo A, Jimenez-Flores R, Laubscher A, Medrano JF. RNA sequencing to study gene expression and single nucleotide polymorphism variation associated with citrate content in cow milk. J Dairy Sci. 2013;96(4):2637–2648. doi: 10.3168/jds.2012-6213.
    1. Maningat PD, Sen P, Sunehag AL, Hadsell DL, Haymond MW. Regulation of gene expression in human mammary epithelium: effect of breast pumping. J Endocrinol. 2007;195(3):503–511. doi: 10.1677/JOE-07-0394.
    1. Maningat PD, Sen P, Rijnkels M, Sunehag AL, Hadsell DL, Bray M, Haymond MW. Gene expression in the human mammary epithelium during lactation: the milk fat globule transcriptome. Physiol Genomics. 2009;37(1):12–22. doi: 10.1152/physiolgenomics.90341.2008.
    1. Lemay DG, Ballard OA, Hughes MA, Morrow AL, Horseman ND, Nommsen-Rivers LA. RNA sequencing of human milk fat layer transcriptome reveals distinct gene expression profiles at three stages of lactation. PLoS ONE. 2013. Published 05 July 2013.
    1. Patton S, Huston GE. Incidence and characteristics of cell pieces on human milk fat globules. Biochim Biophys Acta. 1988;965(2–3):146–153.
    1. Huston GE, Patton S. Factors related to the formation of cytoplasmic crescents on milk fat globules. J Dairy Sci. 1990;73(8):2061–2066. doi: 10.3168/jds.S0022-0302(90)78885-6.
    1. Brenaut P, Bangera R, Bevilacqua C, Rebours E, Cebo C, Martin P. Validation of RNA isolated from milk fat globules to profile mammary epithelial cell expression during lactation and transcriptional response to a bacterial infection. J Dairy Sci. 2012;95(10):6130–6144. doi: 10.3168/jds.2012-5604.
    1. Boutinaud M, Rulquin H, Keisler DH, Djiane J, Jammes H. Use of somatic cells from goat milk for dynamic studies of gene expression in the mammary gland. J Anim Sci. 2002;80(5):1258–1269.
    1. Barboza M, Pinzon J, Wickramasinghe S, Froehlich JW, Moeller I, Smilowitz JT, Ruhaak LR, Huang J, Lonnerdal B, German JB. et al.Glycosylation of human milk lactoferrin exhibits dynamic changes during early lactation enhancing its role in pathogenic bacteria-host interactions. Molecular & cellular proteomics : MCP. 2012;11(6):M111 015248.
    1. Browne EP, Punska EC, Lenington S, Otis CN, Anderton DL, Arcaro KF. Increased promoter methylation in exfoliated breast epithelial cells in women with a previous breast biopsy. Epigenetics. 2011;6(12):1425–1435. doi: 10.4161/epi.6.12.18280.
    1. Monks J, Henson PM. Differentiation of the mammary epithelial cell during involution: implications for breast cancer. J Mammary Gland Biol Neoplasia. 2009;14(2):159–170. doi: 10.1007/s10911-009-9121-0.
    1. Centers for Disease Control and Prevention. Breastfeeding report card-United States, 2010. 2010. [ ]
    1. Hinde K. Richer milk for sons but more milk for daughters: sex-biased investment during lactation varies with maternal life history in rhesus macaques. Am J Hum Biol. 2009;21(4):512–519. doi: 10.1002/ajhb.20917.
    1. VanKlompenberg MK, McMicking HF, Hovey RC. Technical note: a vacuum-assisted approach for biopsying the mammary glands of various species. J Dairy Sci. 2012;95(1):243–246. doi: 10.3168/jds.2011-4565.
    1. Hartono S. Globulator. 2013. [ ]
    1. Collins TJ. ImageJ for microscopy. Biotechniques. 2007;43(1 Suppl):25–30.
    1. UC Davis-Bioinformatics. 2012. [ ]
    1. UC Davis-Bioinformatics. 2013. [ ]
    1. Babraham Bioinformatics. 2012. [ ]
    1. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10(3):R25. doi: 10.1186/gb-2009-10-3-r25.
    1. Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25(9):1105–1111. doi: 10.1093/bioinformatics/btp120.
    1. Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, Salzberg SL, Wold BJ, Pachter L. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;28(5):511–515. doi: 10.1038/nbt.1621.
    1. Flicek P, Amode MR, Barrell D, Beal K, Brent S, Carvalho-Silva D, Clapham P, Coates G, Fairley S, Fitzgerald S. et al.Ensembl 2012. Nucleic Acids Res. 2012;40(Database issue):D84–D90.
    1. NCBI Resource Coordinators. Database resources of the National Center for Biotechnology Information. Nucleic Acids Res. 2013;41(Database issue):D8–D20.
    1. NCBI Gene Expression Omnibus. 2013. [ ]
    1. Anders S. HTSeq: Analysing high-throughput ssequencing data with Python. 2013. [ ]
    1. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25(16):2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. Anders S, Huber W. Differential expression analysis for sequence count data. Genome Biol. 2010;11(10):R106. doi: 10.1186/gb-2010-11-10-r106.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Stat Soc B Met. 1995;57(1):289–300.
    1. da Huang W, Sherman BT, Tan Q, Collins JR, Alvord WG, Roayaei J, Stephens R, Baseler MW, Lane HC, Lempicki RA. The DAVID Gene Functional Classification Tool: a novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 2007;8(9):R183. doi: 10.1186/gb-2007-8-9-r183.
    1. Taylor-Papadimitriou J, Stampfer M, Bartek J, Lewis A, Boshell M, Lane EB, Leigh IM. Keratin expression in human mammary epithelial cells cultured from normal and malignant tissue: relation to in vivo phenotypes and influence of medium. J Cell Sci. 1989;94(Pt 3):403–413.
    1. Greenberg AS, Egan JJ, Wek SA, Garty NB, Blanchette-Mackie EJ, Londos C. Perilipin, a major hormonally regulated adipocyte-specific phosphoprotein associated with the periphery of lipid storage droplets. J Biol Chem. 1991;266(17):11341–11346.
    1. Russell TD, Palmer CA, Orlicky DJ, Fischer A, Rudolph MC, Neville MC, McManaman JL. Cytoplasmic lipid droplet accumulation in developing mammary epithelial cells: roles of adipophilin and lipid metabolism. J Lipid Res. 2007;48(7):1463–1475. doi: 10.1194/jlr.M600474-JLR200.
    1. Chong BM, Reigan P, Mayle-Combs KD, Orlicky DJ, McManaman JL. Determinants of adipophilin function in milk lipid formation and secretion. Trends Endocrinol Metab. 2011;22(6):211–217. doi: 10.1016/j.tem.2011.04.003.
    1. Chong BM, Russell TD, Schaack J, Orlicky DJ, Reigan P, Ladinsky M, McManaman JL. The adipophilin C terminus is a self-folding membrane-binding domain that is important for milk lipid secretion. J Biol Chem. 2011;286(26):23254–23265. doi: 10.1074/jbc.M110.217091.
    1. Russell TD, Schaack J, Orlicky DJ, Palmer C, Chang BH, Chan L, McManaman JL. Adipophilin regulates maturation of cytoplasmic lipid droplets and alveolae in differentiating mammary glands. J Cell Sci. 2011;124(Pt 19):3247–3253.
    1. McIntosh AL, Senthivinayagam S, Moon KC, Gupta S, Lwande JS, Murphy CC, Storey SM, Atshaves BP. Direct interaction of Plin2 with lipids on the surface of lipid droplets: a live cell FRET analysis. Am J Physiol Cell Physiol. 2012;303(7):C728–C742. doi: 10.1152/ajpcell.00448.2011.
    1. Chambers JM, Cleveland WS, Tukey PA, Kleiner B. Graphical Methods for Data Analysis. Boston, Massachusetts: Duxbury Press; 1983.
    1. Boutinaud M, Jammes H. Potential uses of milk epithelial cells: a review. Reprod Nutr Dev. 2002;42(2):133–147. doi: 10.1051/rnd:2002013.
    1. Gaffney EV, Polanowski FP, Blackburn SE, Lambiase JP. Origin, concentration and structural features of human mammary gland cells cultured from breast secretions. Cell and tissue research. 1976;172(2):269–279.
    1. Department of Health and Human Services. The Surgeon General’s Call to Action to Support Breastfeeding. Washington, DC: U.S. Department of Health and Human Services, Office of the Surgeon General; 2011.
    1. Fung C, Lock R, Gao S, Salas E, Debnath J. Induction of autophagy during extracellular matrix detachment promotes cell survival. Mol Biol Cell. 2008;19(3):797–806.
    1. Schedin P. Pregnancy-associated breast cancer and metastasis. Nat Rev Cancer. 2006;6(4):281–291. doi: 10.1038/nrc1839.
    1. Bladstrom A, Anderson H, Olsson H. Worse survival in breast cancer among women with recent childbirth: results from a Swedish population-based register study. Clin Breast Cancer. 2003;4(4):280–285. doi: 10.3816/CBC.2003.n.033.
    1. Stensheim H, Moller B, van Dijk T, Fossa SD. Cause-specific survival for women diagnosed with cancer during pregnancy or lactation: a registry-based cohort study. J Clin Oncol: Official Journal of the American Society of Clinical Oncology. 2009;27(1):45–51.
    1. Johansson AL, Andersson TM, Hsieh CC, Cnattingius S, Lambe M. Increased mortality in women with breast cancer detected during pregnancy and different periods postpartum. Cancer Epidem Biomar: A Publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2011;20(9):1865–1872.
    1. Zhou Q, Li M, Wang X, Li Q, Wang T, Zhu Q, Zhou X, Gao X, Li X. Immune-related microRNAs are abundant in breast milk exosomes. Int J Biol Sci. 2012;8(1):118–123.
    1. Munch EM, Harris RA, Mohammad M, Benham AL, Pejerrey SM, Showalter L, Hu M, Shope CD, Maningat PD, Gunaratne PH. et al.Transcriptome profiling of microRNA by Next-Gen deep sequencing reveals known and novel miRNA species in the lipid fraction of human breast milk. PLoS ONE. 2013;8(2):e50564. doi: 10.1371/journal.pone.0050564.
    1. Cline JM, Wood CE. The mammary glands of macaques. Toxicol Pathol. 2008;36(7):134s–141s.
    1. O’Sullivan A, He X, McNiven EM, Hinde K, Haggarty NW, Lonnerdal B, Slupsky CM. Metabolomic phenotyping validates the infant rhesus monkey as a model of human infant metabolism. J Pediatr Gastroenterol Nutr. 2013;56(4):355–363. doi: 10.1097/MPG.0b013e31827e1f07.
    1. O’Sullivan A, He X, McNiven EM, Haggarty NW, Lonnerdal B, Slupsky CM. Early diet impacts infant rhesus gut microbiome, immunity, and metabolism. J Proteome Res. 2013;12(6):2833–2845. doi: 10.1021/pr4001702.

Source: PubMed

3
订阅