Publications

Here is a selection of publications where different laminin isoforms were used to create more authentic cell culture systems.

  • Sort by

  • Area of interest

  • Toward Xeno-Free Differentiation of Human Induced Pluripotent Stem Cell-Derived Small Intestinal Epithelial Cells

    Jaakko Saari, Fatima Siddique, Sanna Korpela, Elina Mäntylä, Teemu O. Ihalainen, Katri Kaukinen, Katriina Aalto-Setälä, Katri Lindfors, and Kati Juuti-Uusitalo. International Journal of Molecular Sciences, 2022

    Small intestinal epithelial cells were differentiated from induced pluripotent stem cells (iPSC-SIEC), via definitive endoderm, on Biolaminin 511. The aim of the study was to develop a protocol with animal-free components to reduce variation in experimental outcomes. The differentiated cells cultured on laminin exhibited more enterocyte specific cellular functionality than cells on Geltrex or collagen. The protocol is serum-free and can be easily modified as completely xeno-free by culturing iPSCs on Biolaminin 521 matrix and changing the B27 and N2 supplement to available xeno-free versions.

  • Laminin as a Potent Substrate for Large-Scale Expansion of Human Induced Pluripotent Stem Cells in a Closed Cell Expansion System

    Gjorevski N. & Lutoff MP. Nat Protoc, 2017

    In this work, the authors describe a protocol for the generation of well-defined matrices for the culture of intestinal stem cells (ISCs) and intestinal organoids, using PEG hydrogel backbone functionalized with minimal adhesion cues including RGD (Arg-Gly-Asp), which is sufficient for ISC expansion, and laminin-111, which is required for organoid formation.

  • Designer matrices for intestinal stem cell and organoid culture

    Gjorevski N., Sachs N., Manfrin A., Giger S., Bragina M.E., Ordóñez-Morán P., Clevers H., Lutolf M.P. Nature letters, 2016

    Here the authors used modular synthetic hydrogel (cross-linked poly(ethylene glycol) (PEG)) to define the key extracellular matrix (ECM) parameters that govern intestinal stem cell (ISC) expansion and organoid formation, and show that separate stages of the process require different mechanical environments and ECM components. Fibronectin-based adhesion was sufficient for ISC survival and proliferation and high matrix stiffness significantly enhanced ISC expansion through a yes-associated protein 1 (YAP)-dependent mechanism. ISC differentiation and organoid formation, on the other hand, required a soft matrix and full-length laminin-111-based adhesion. The authors also produced mechanically dynamic matrices that were initially optimal for ISC expansion and subsequently permissive to differentiation and intestinal organoid formation.

  • Abnormal Wnt and PI3Kinase Signaling in the Malformed Intestine of lama5 Deficient Mice

    Ritié L., Spenlé C., Lacroute J.I., Bolcato-Bellemin A-L., Lefebvre O., Bole-Feysot C., Jost B., Klein A., Arnold C., Kedinger M., Bagnard D., Orend G., Simon-Assmann P. PLoS One, 2012

    Laminin-511 is highly expressed in the intestine. To understand the mechanistic role of laminin-511 in tissue homeostasis, the researchers used RNA profiling of embryonic intestinal tissue of lama5 knockout mice and identified a lama5 specific gene expression signature. They show that laminin a5 plays a crucial role in both epithelial and mesenchymal (smooth muscle) cell behavior by inhibiting Wnt and activating PI3K signaling. We conclude that conflicting signals are elicited in the absence of lama5, which alter cell adhesion, migration as well as epithelial and muscle differentiation. The LMa5 deficient intestine also displays a smooth muscle defect and myogenic differentiation markers are affected. Laminin-511 supports adhesion of epithelial cells and Akt phosphorylation. Laminin-511 stimulates the spreading of epithelial and muscle cells (compared to laminin-111). Inhibition of Akt with wortmannin abolished spreading of epithelial cells on laminin-511 as evidenced by cell laminin-511 specifically activates Akt through the PI3K pathway in intestinal epithelial but not in mesenchymal cells. Cell migration was also higher on Laminin-511. Laminin-511 also protects cells against H2O2-induced apoptosis.

  • Laminins in the Developing and Adult Human Small Intestine: Relation With the Functional Absorptive Unit

    Teller I.C., Auclair J., Herring E., Gauthier R., Ménard D., Beaulieu J-F.Developmental dynamics, 2007

    Here, the expression of the five laminin a-chains was analyzed in the developing and mature human small intestine at the protein and transcript levels in order to further delineate specific involvement of individual laminins in relation to the epithelial cell state as defined along the functional crypt-villus axis. The results show that all of the a-laminins are expressed in significant amounts in the small intestine relative to a panel of other tissues and organs. Distinct epithelial and mesenchymal origins, as well as a differential occurrence in intestinal basement membranes according to developmental stage, along the crypt-villus axis and in compartment-related experimental intestinal cell models. Taken together, the data point out the prime importance of a2-, a3-, and a5-containing laminins for the development and maintenance of the functional human intestinal epithelium.

  • Laminin α5 influences the architecture of the mouse small intestinal mucosa

    Mahoney Z.X., Stappenbeck T.S., Miner J.HJ Cell Sci. 2008

    The villus basement membrane is rich in laminin α5. Here the authors show that diminution of laminin α5 in a mouse model led to a compensatory deposition of colonic laminins that resulted in a transformation from a small intestinal to a colonic mucosal architecture. The alteration in mucosal architecture was associated with reduced levels of nuclear p27Kip1, a cell cycle regulator, and altered intestinal epithelial cell proliferation, migration, and differentiation. The results suggest that laminin α5 plays a crucial role in establishing and maintaining the specific mucosal pattern of the mouse small intestine.

  • Laminin a5 chain is required for intestinal smooth muscle development

    Bolcato-Bellemin A-L., Lefebvre O., Arnold C., Sorokin L., Miner J. H., Kedinger M., Simon-Assmann P. Developmental Biology, 2003

    Here, the function of the laminin a5 chain in the developing intestine was defined by analyzing laminin a5 -/- mutants and by grafting experiments. The authors show that laminin a5 plays a major role in smooth muscle organization and differentiation, as excessive folding of intestinal loops and delay in the expression of specific markers are observed in laminin a5 -/- mice. Loss of a5 expression was paralleled by ectopic or accelerated deposition of laminin a2 and a4 chains; this may explain why no obvious defects were observed in the villous form and enterocytic differentiation. Lack of the laminin a5 chain was accompanied by a decrease in epithelial a3B1 integrin receptor expression adjacent to the epithelial basement membrane and of Lutheran blood group glycoprotein in the smooth muscle cells, indicating that these receptors are likely mediating the a5 interactions. Taken together, the laminin a5 chain is essential for the normal development of the intestinal smooth muscle.

  • L1CAM defines the regenerative origin of metastasis-initiating cells in colorectal cancer

    Ganesh K., Basnet H., Kaygusuz Y., Laughney A.M., He L., Sharma R., O’Rourke K.P., Reuter V.P., Huang Y.-H., Turkekul M., Er E.E., Masilionis I., Manova-Todorova K., Weiser M.R., Saltz L.B., Garcia-Aguilar J., Koche R., Lowe S.W., Pe’er D., Shia J., Massagué J.Nature Cancer, 2020

    The authors show that L1CAM+ cells in human colorectal cancer (CRC) have metastasis-initiating capacity, and they define their relationship to tissue regeneration. By using recombinant L1CAM extracellular domain and basement membrane components, they confirmed that L1CAM bound heterophilically to laminins known to be expressed in the intestinal and endothelial cell basement membranes in addition to exhibiting homophilic interaction with L1CAM itself. L1CAM knockdown inhibited the ability of CRC organoid-derived cells to bind to laminin-coated plates. Together, these data suggest that L1CAM enables the adhesion of metastasis-initiating cells to laminin-rich basement membranes, which is required for metastasis and organoid growth.

  • Tissue distribution of the laminin β1 and β2 chain during embryonic and fetal human development

    Roediger M., Miosge N., Gersdorff N. J Mol Hist., 2010

    Here, the authors investigated the tissue distribution of the laminin b1 and b2 chains on the protein level in various developing embryonic and fetal human organs between gestational weeks 8 and 12. The laminin b1 chain was ubiquitously expressed in the basement membrane zones of the brain, ganglia, blood vessels, liver, kidney, skin, pancreas, intestine, heart and skeletal system. Furthermore, the laminin b2 chain was present in the basement membrane zones of the brain, ganglia, skin, heart, and skeletal system. The findings of this study support and expand upon the theory that these two laminin chains are important during human development. In cartilage, the laminin b1 chain was expressed from gw 10 onwards but not during gw 8 and 9, whereas the detection of the laminin b2 chain was limited to gw 8 and 9. This indicates a developmental switch in the laminin b chain and suggests that the laminin b1 chain does not play a role in human cartilage development until the fetal stage. In human fetal cartilage (gw 17 and 24), a strong pericellular immunohistochemical reaction for laminin 111 was shown. In embryo chick sternum and mouse limb bud, laminin b1 and b2 chains are present in the cytoplasm of chondrocytes.

  • Immunohistochemical Distribution of Laminin-5 γ 2 Chain and its Developmental Change in Human Embryonic and Foetal Tissues

    Lu W., Miyazaki K., Mizushima H., Nemoto N.

    Here, immunohistochemical distribution of laminin γ2 chain, a subunit of the basement membrane protein laminin 332, was examined in 19 cases of human embryos and foetuses ranging from 4 to 25 weeks of gestation. Laminin γ2 was first detected in the basement membranes underlying ectodermal epithelial tissues, such as the skin and tooth, as early as 5–6 weeks of gestation. Between 6–7 and 12–13 weeks, laminin γ2 was detected in the basement membranes of various endodermal epithelial tissues, such as the bronchus, oesophagus, stomach, intestines, urinary bladder, gallbladder, and hepatopancreatic duct. The deposition of laminin γ2 in the basement membrane was associated with the process of morphogenesis. In the small intestine, laminin γ2 first appeared in the basement membrane of the primitive short villi, and its level gradually increased in the villus region but decreased in the cryptic region during the maturation of the organ. In addition, non-basement membrane immunoreactivity for laminin γ2 was detected in some mesoderm-derived tissues, such as the cartilage and skeletal and smooth muscle fibers. These results suggest a common role of laminin 332 and some specific roles of its γ2 chain in the morphogenesis of human tissues.