• Ei tuloksia

The aim of this Master’s thesis was to investigate the role of LMα5 in MG development, as it has not been researched before. MG development is an intricate process involving an interplay of several systemic and local factors.

The investigations in this thesis were divided into in vitro and in vivo methods. The in vitro methods involved analysis of growth rates and structures of organoids grown from isolated MMECs, and the in vivo methods explored the development and structure of the mammary tree in isolated MGs.

According to the findings of this thesis, LMα5 is a key player in MG development, as witnessed by the in vitro branching assay and in vivo experiments. Branching morphogenesis is stunted in LAMA5 knockout cells and mice. Organoids grown from LAMA5 deficient cells form fewer branches than their wild type counterparts. In MGs isolated from mice lacking LMα5, both epithelial length and number of TEB structures, key points for branching morphogenesis, are reduced. On closer inspection of epithelial structure, disorganized luminal assembly can be observed, with detached aggregates of cells in the ductal lumen. This can be a sign of loss of luminal epithelial cell contact with the BM, as luminal cells normally are in charge of LMα5 deposition into the BM. Loss of BM contact can also result in disturbed apico-basal polarity, an event witnessed in other LMα5 deficient tissues researched before. This deduction is fortified by related studies where loss of β1 integrin, a mediator of laminin signaling, has been shown to result in defective cellular polarity. Together, these findings indicate a disturbance in normal MG development and tissue remodeling in LMα5 deficient MGs.

The experiments performed for this thesis lay a groundwork for further studies on the significance of LMα5, and other understudied laminins, on MG development. As LMα5 has been shown to enforce luminal expression of epithelial cells, it would be interesting to explore its effects throughout MG development, beginning from MaSC lineage re-striction in the embryo. Future studies can also concentrate on more detailed investigations on the mechanisms behind the phenomena displayed in pubertal and pregnant LMα5 knockout glands.

In addition to significance in developmental biology, the presented findings can have a consideration in future MG tissue engineering applications. In the tissue engineering

triad, scaffolds are often described as the supportive framework on which tissues and organs can be built. They provide cues for cellular proliferation and differentiation. ECMs and BMs are nature’s own scaffolds, working in concert with cells to build functional units.

In MG, this concert is in discord when LMα5 is removed.

REFERENCES

Akhtar, N. & Streuli, C.H. (2012). An integrin-ILK-microtubule network orients cell polarity and lumen formation in glandular epithelium. Nature Cell Biology, Volume 15, Number 1, January 2012, pp. 17-27.

Aumalley, M. (2013). The laminin family. Cell Adhesion & Migration 7:1, 48–55;

January/February 2013.

Ball, S.M. (1998). The Development of the Terminal End Bud in the Prepubertal-Pubertal Mouse Mammary Gland. The Anatomical Record 250:459-464 (1998), pp.459-464.

Bouabe, H. & Okkenhaug, K. (2013). Gene Targeting in Mice: a Review. Methods Mol Biol. 2013 ; 1064: 315–336.

Carter, M. & Shieh, J.C. (2010). Guide to Research Techniques in Neuroscience.

Academic Press, Elsevier Inc., pp. 281-296.

Corning (2019). Corning® Matrigel® Matrix, web page. Available (accessed 11.12.2019):

https://www.corning.com/worldwide/en/products/life-sciences/products/surfaces/matrigel-matrix.html

Cowin, P. & Wysolmerski, J. (2010). Molecular Mechanisms Guiding Embryonic Mammary Gland Development. Cold Spring Harb Perspect Biol 2010;2:a003251 Dahl-Jensen, S. & Grapin-Botton, A. (2017). The physics of organoids: a

biophysical approach to understanding organogenesis. Development (2017) 144, 946-951.

Doi, M. et al. (2002). Recombinant Human Laminin-10 (α5β1γ1). The Journal of Biological Chemistry, Vol. 277, No. 15, Issue of April 12, pp. 12741-12748.

Dos Santos, C.O. et al. (2015). An epigenetic memory of pregnancy in the mouse mammary gland. Cell Rep. 2015 May 19;11(7):1102-9.

Durbeej, M. (2010). Laminins. Cell Tissue Res (2010) 339:259–268.

Eisenstein, M. (2019). Giving organoids room to grow. Lab Animal, Vol 48, July 2019, 193-195.

Elo, T., Lindfors, P.H., Lan, Q. et al. (2017). Ectodysplasin target gene Fgf20 regulates mammary bud growth and ductal invasion and branching during puberty. Sci Rep 7, 5049 (2017).

Englund, J.I. et al. (unpublished). Laminin alpha 5 is Necessary for Mammary Ep-ithelial Growth and Function by Maintaining Luminal EpEp-ithelial Cell Identity. Avail-able (accessed 09.02.2020): https://doi.org/10.1101/2019.12.27.889451

Ewald, A.J. et al. (2008). Collective Epithelial Migration and Cell Rearrangements Drive Mammary Branching Morphogenesis. Dev Cell, 2008 April, 14(4): pp. 570-581.

Feil, S., Valtcheva, N. & Feil, R. (2009). Inducible Cre Mice. In Ralf Kühn, Wolfgang Wurst (Eds.), Gene Knockout Protocols: Second Edition, vol. 530 (pp.

343-363), Humana Press, a part of Springer Science+Business Media, LLC 2009.

Furuta, S. & Bissell, M.J. (2016). Pathways Involved in Formation of Mammary Organoid Architecture Have Keys to Understanding Drug Resistance and to Discovery of Druggable Targets. Cold Spring Harbor Symposia on Quantitative Biology, Volume LXXXI.

Ghajar, C.M. & Bissell, M.J. (2008). Extracellular matrix control of mammary gland morphogenesis and tumorigenesis: insights from imaging. Histochem Cell Biol (2008) 130:1105-1118.

Goodwin, L.D. & Leech, N.L. (2006). Understanding Correlation: Factors That Affect the Size of r. The Journal of Experimental Education, 2006, 74(3), 251-266.

Guldager Kring Rasmussen, D. & Karsdal, M.A. (2016). Laminins. In Morten A.

Karsdal, Diana J. Leeming, Kim Henriksen & Anne-Christine Bay-Jensen (Eds.), Biochemistry of Collagens, Laminins and Elastin. Structure, Function and

Biomarkers (pp. 163-187). Academic Press.

Halfter, W. et al. (2015). New concepts in basement membrane biology. FEBS Journal 282 (2015) 4466-4479.

Herrmann, H. et al. (2007). Intermediate filaments: from cell architecture to nanomechanics. Nat Rev Mol Cell Biol 8, 562–573 (2007).

Hinck, L. & Silberstein, G.B. (2005). Key stages in mammary gland development.

The mammary end bud as a motile organ. Breast Cancer Research 2005, 7:245-251.

Hohenester, E. & Yurchenco, P.D. (2013). Laminins in basement membrane assembly. Cell Adhesion & Migration 7:1, 56-63; January/February 2013.

Howard, B.A. (2012). In the beginning: The establishment of the mammary lineage during embryogenesis. Seminars in Cell & Developmental Biology 23 (2012) 574-582.

Inman, J.L. et al. (2015). Mammary gland development: cell fate specification, stem cells and the microenvironment. Development (2015) 142, pp. 1028-1042.

Jamieson, P.R. et al. (2017). Derivation of a robust mouse mammary organoid system for studying tissue dynamics. Development (2017) 144, 1065-1071.

Jardé, T. et al. (2016). Wnt and Neuregulin1/ErbB signalling extends 3D culture of hormone responsive mammary organoids. Nature Communications, 7:13207.

Jayadev, R. & Sherwood, D.R. (2017). Basement membranes. Current Biology 27, R199-R217, March 20, 2017.

Jewett, C.E. & Prekeris, R. (2018). Insane in the apical membrane: Trafficking events mediating apicobasal epithelial polarity during tube morphogenesis. Traffic 2018;19:666-678.

Kikkawa, Y. et al. (2017). Identification of laminin α5 short arm peptides active for endothelial cell attachment and tube formation. J.Pept.Sci. 2017; 23: 666-673.

Kwan, K-M. (2002). Conditional Alleles in Mice: Practical Considerations for Tissue-Specific Knockouts. Genesis 32:49-62 (2002).

LaMarca, H.L. & Rosen, J.M. (2007). Estrogen regulation of mammary gland development and breast cancer: amphiregulin takes center stage. Breast Cancer Research 2007, 9:304.

Macherey-Nagel (2017). User manual, NucleoSpin® Tissue. Januar 2017 / Rev.

17.

Macias, H. & Hinck, L. (2012). Mammary Gland Development. Wiley Interdiscip Rev Dev Biol. 2012; 1(4): 533-557.

Mailleux, A.A., Overholtzer, M. & Brugge, J.S. (2008). Lumen formation during mammary epithelial morphogenesis. Insights from in vitro and in vivo models.

Cell Cycle 7:1, 57-62.

Maizels, N. (2013). Genome Engineering with Cre-loxP. J Immunol 2013; 191:5-6.

Maller, O., Martinson, H. & Schedin, P. (2010). Extracellular Matrix Composition Reveals Complex and Dynamic Stromal-Epithelial Interactions in the Mammary Gland. J Mammary Gland Biol Neoplasia (2010) 15:301–318.

McNally, S. & Martin, F. (2011). Molecular regulators of pubertal mammary gland development. Annals of Medicine, 2011; 43: 212-234.

Mroue, R. & Bissell, M.J. (2012). Three-Dimensional Cultures of Mouse Mammary Epithelial Cells. In Scott H. Randell and M. Leslie Fulcher (Eds.),

Epithelial Cell Culture Protocols: Second Edition, Methods in Molecular Biology, vol. 945 (pp. 221-250), ©Springer Science+Business Media, LLC 2012.

Muthuswamy, S.K. et al. (2001). ErbB2, but not ErbB1, reinitiates proliferation and induces luminal repopulation in epithelial acini. Nature Cell Biology, Vol. 3, September 2001.

Paine, I.S. & Lewis, M.T. (2017). The Terminal End Bud: the Little Engine that Could. J Mammary Gland Biol Neoplasia (2017) 22:93-108.

Perry, J.K. et al. (2008). The contribution of growth hormone to mammary neoplasia. J Mammary Gland Biol Neoplasia. 2008 Mar; 13(1): 131–145.

Petersen, O.W. et al. (1992). Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proc. Natl. Acad. Sci. USA, Vol. 89, pp. 9064-9068, October 1992.

Pouliot, N. & Kusuma, N. (2013). Laminin-511. A multi-functional adhesion protein regulating cell migration, tumor invasion and metastasis. Cell Adhesion &

Migration 7:1, 142-149; January/February 2013.

Pozzi, A., Yurchenco, P.D. & Iozzo, R.V. (2017). The nature and biology of basement membranes. Matrix Biol. (2017) 57-58, 1-11.

Raouf, A. et al. (2012). The biology of human breast epithelial progenitors.

Seminars in Cell & Developmental Biology 23 (2012) 606-612.

Richert, M.M. et al. (2000). An Atlas of Mouse Mammary Gland Development.

Journal of Mammary Gland Biology and Neoplasia, Vol. 5., No. 2, 2000, pp. 227-231.

Rillema, J. A. (2014). Mammary gland. In AccessScience. McGraw-Hill Education. Available (accessed 10.12.2019):

https://doi-org.libproxy.tuni.fi/10.1036/1097-8542.402600

Ross, M.H. & Pawlina, W. (2016). Histology: A Text and Atlas. With Correlated Cell and Molecular Biology (7th ed.), Wolters Kluwer Health, Philadelphia, pp.

866-872.

Sampayo, R. et al. (2018). Mammary Gland Organoids. In S. Soker, A. Skardal (Eds.), Tumor Organoids, Cancer Drug Discovery and Development (pp. 35-49).

Springer International Publishing AG 2018.

Sasaki, T., Fässler, R. & Hohenester, E. (2004). Laminin: the crux of basement membrane assembly. The Journal of Cell Biology, Volume 164, Number 7, March 29, 2004 959-963.

Slade, M.J. et al. (1999). The Human Mammary Gland Basement Membrane Is Integral to the Polarity of Luminal Epithelial Cells. Experimental Cell Research 247, 267-278 (1999).

Smith, G.H. (2005). Label-retaining epithelial cells in mouse mammary gland di-vide asymmetrically and retain their template DNA strands. Development 2005 132: 681-687.

Spenlé, C. et al. (2013). Laminin α5 guides tissue patterning and organogenesis.

Cell Adhesion & Migration 7:1, 90-100; January/February 2013, pp. 90-100.

The Jackson Laboratory (n.d.). STOCK Tg(Krt8-cre/ERT2)17Blpn/J, web page.

Available (accessed 11.12.2019): https://www.jax.org/strain/017947

Tortora, G.J. & Derrickson, B. (2007). Principles of Anatomy and Physiology (11th ed.), John Wiley & Sons, Inc, pp. 1083-1084.

Van Keymeulen, A. et al. (2011) Distinct stem cells contribute to mammary gland development and maintenance. Nature, Vol. 479, 10 November 2011, pp. 189-193.

Vector Biolabs (n.d.). AAV-Cre (AAV serotype 2) AAV, web page. Available (accessed 13.12.2019): https://www.vectorbiolabs.com/product/7011-aav-cre-aav-serotype-2-aav/

Vector Biolabs (n.d.). AAV-Cre (AAV serotype 9) AAV, web page. Available (accessed 13.12.2019): https://www.vectorbiolabs.com/product/7014-aav-cre-aav-serotype-9-aav/

Wang, F. et al. (2012). RNAscope®: A Novel In Situ RNA Analysis. Platform for Formalin-Fixed Paraffin-Embedded Tissues. J. Mol. Diagnostics, 2012, 14:22–29.

Watson, C.J. & Khaled, W.T. (2008). Mammary development in the embryo and adult: a journey of morphogenesis and commitment. Development 135, pp. 995-1003 (2008).

Wuidart, A. et al. (2018). Early lineage segregation of multipotent embryonic mammary gland progenitors. Nature Cell Biology, Vol. 20, June 2018, pp. 666-676.

Yao, Y. (2017). Laminin: loss-of-function studies. Cell. Mol. Life Sci. (2017) 74:1095-1115.

Young, B., Woodford, P. & O’Dowd, G. (2014). Wheater’s Functional Histology. A Text and Colour Atlas (6th ed.), Elsevier Churchill Livingstone, p. 378.

Yurchenco, P.D. (2011). Basement Membranes: Cell Scaffoldings and Signaling Platforms. Cold Spring Harb Perspect Biol 2011;3:a004911.