18 research outputs found

    Functional living biointerfaces to direct cell-material interaction

    Full text link
    [EN] This thesis deals with the development of a living biointerface between synthetic substrates and living cells to engineer cell-material interactions for tissue engineering purposes. This living biointerface is made of Lactococcus lactis, a non-pathogenic lactic bacteria widely used as starter in the dairy industry and, recently, in the expression of heterologous proteins in applications such as oral vaccine delivery or membrane-bound expression of proteins. L. lactis has been engineered to display the III 7-10 fragment of the fibronectin fused to GFP as reporter protein. Fibronectin is a ubiquitous protein present in the extracellular matrix, a complex mesh of structural and adhesive proteins which serve as mechanical support and development niche for cells of a wide variety of tissues. This fragment contains two important sequences, RGD and PHSRN. RGD is an adhesive sequence that interacts with a wide range of integrins, membrane-bound receptors that play a role in cellular processes such as adhesion, migration, proliferation and differentiation. On the other hand, PHSRN binds synergistically with RGD to some integrins such as alpha-5-beta-1 and others, increasing the specificity of this interaction. Genetically engineered L. lactis has been thoroughly characterized to test its capabilities as a living interface. This strain was found to express the FNIII 7-10-GFP fragment covalently linked to the cell wall and biological activity and expression levels of this fragment was assessed with techniques such as Western blot, ELISA and immunofluorescence. Moreover, this strain still holds the ability to develop biofilms, communities of sessile, attached bacteria to abiotic surfaces which helps greatly in the generation of a stable monolayer of bacteria between synthetic substrates and mammalian cells. Mammalian cell behaviour in response to the expressed fibronectin fragment on L. lactis membrane was also assessed. Several cell lines were tested, such as Fn-/Fn- and NIH3T3 fibroblasts, C2C12 myoblasts and human bone-marrow derived mesenchymal cells. This living biointerface was found to trigger cell adhesion and FAK phosphorylation, a marker for intracellular integrin-mediated signalling in all of the tested cell lines. It also triggered myoblast-to-myotube differentiation on C2C12 cells. In hMSCs, the cell-wall exposed fibronectin fragment was found to enhance the phosphorylation of ERK1/2, a kinase involved in the MAPK pathway, which is deeply involved in a multitude of cellular processes related to differentiation, proliferation and migration. Nevertheless, this thesis is a proof of concept that this novel system can be further exploited to express almost any desired protein or small molecule to help in the development of new tissues from progenitor cells. These molecules can be either secreted in the medium or displayed in the membrane, and can also be constitutively expressed or in-demand, due to the great flexibility of L. lactis and the wide variety of expression systems available. This interface based on living bacteria establishes a new paradigm in surface functionalization for biomedical engineering applications.[ES] Esta tesis aborda el desarrollo de una biointerfase viviente entre materiales sintéticos y células vivas con el objetivo de dirigir la interacción célula-material en aplicaciones de ingeniería tisular. Esta biointerfase está compuesta de Lactococcus lactis, una bacteria láctica no patógena, ampliamente usada en la industria láctea como inóculo, y, recientemente, en la expresión heteróloga de proteínas para su uso como vacunas de administración oral o su expresión en membrana. L. lactis ha sido genéticamente modificado para expresar el fragmento III 7-10 de la fibronectina, unida a GFP como reporter. La fibronectina es una proteína presente de forma ubicua en la matriz extracelular, una compleja red de proteínas adhesivas y estructurales cuyo propósito es servir como soporte estructural y como nicho de desarrollo para diversos tejidos. Este fragmento contiene dos secuencias importantes, RGD y PHSRN. RGD es una secuencia adhesiva de unión que interacciona con una amplia variedad de integrinas, receptores de membrana que juegan muchos e importantes papeles en diferentes procesos celulares, como adhesión, proliferación, migración o diferenciación. Por otra parte, PHSRN se une a las integrinas de forma sinérgica con RGD facilitando aún más estos procesos y aumentando la especificidad de esta interacción. Esta cepa de L. lactis modificada ha sido ampliamente caracterizada para estudiar su idoneidad como interfaz funcional viviente. Se ha demostrado que L. lactis es capaz de expresar el fragmento FNIII7-10-GFP covalentemente anclado a la pared celular bacteriana, habiéndose caracterizado también su actividad biológica con técnicas como Western blot, ELISA e inmunofluorescencia. Esta cepa mantiene la capacidad de desarrollo de biofilms presente en la gran mayoría de microorganismos. Los biofilms son comunidades de bacterias sésiles adheridas a un sustrato que pueden ser usadas como interfase física entre células de mamífero y sustratos abióticos. También se ha estudiado la respuesta celular a la fibronectina expuesta en la membrana de L. lactis. Se estudiaron varias líneas celulares, como fibroblastos Fn-/Fn- y NIH3T3, mioblastos C2C12 y células mesenquimales humanas derivadas de médula ósea. Esta interfase viviente fue capaz de provocar respuesta celular en forma de adhesión en todas las líneas estudiadas, además de inducir diferenciación de mioblastos a miotubos en C2C12 y de provocar la fosforilación de FAK, un marcador de señalización celular mediada por integrinas. En células mesenquimales humanas se demostró la capacidad del fragmento de fibronectina expuesto para fosforilar ERK1/2, una kinasa perteneciente a la ruta de señalización MAPK, ruta que forma parte de muchos procesos celulares importantes como diferenciación, proliferación y migración. Pese a todo, esta tesis es sólo una prueba de concepto de un sistema que puede ser utilizado para expresar casi cualquier proteína o molécula pequeña deseada, que puede ser muy útil en el desarrollo de nuevos tejidos a partir de sus células progenitoras. Estas moléculas pueden ser secretadas en el medio o ancladas en la pared celular, de forma constitutiva o bajo demanda, debido a la flexibilidad y amplia variedad de sistemas de expresión disponibles para L. lactis. Esta biointerfase basada en bacterias vivas establece un nuevo paradigma en el campo de la funcionalización de superficies para aplicaciones de ingeniería biomédica.[CA] Aquesta tesi aborda el desenvolupament d'una interfase viva entre materials sintètics i cèl·lules vives amb l'objectiu de dirigir la interacció cèl·lula-material, per al seu ús en aplicacions d'enginyeria tissular. Aquesta interfase està composta de Lactococcus lactis, un bacteri làctic, no patogènic i àmpliament utilitzat en l'industria làctica com a inòcul, i, recentment, en l'expressió heteròloga de proteïnes per al seu ús com vacunes d'administració oral o per a la seva expressió en membrana. L. lactis ha sigut genèticament modificada per a expressar el fragment III7-10 de la fibronectina, unida a GFP com a reporter. La fibronectina és una proteïna present de forma ubiqua en la matriu extracel·lular, una complexa xarxa de proteïnes adhesives i estructurals que s'utilitzen com a suport estructural i com a nínxol de desenvolupament per a diversos teixits. Aquest fragment conté dos seqüències importants, RGD i PHSRN. RGD és una seqüència adhesiva d'unió a integrines, receptors de membrana que juguen molts i molt importants papers en diferents processos cel·lulars, com poden ser adhesió, proliferació, migració o diferenciació. Per altra banda, PHSRN s'uneix a les integrines de forma sinèrgica amb RGD facilitant encara més aquests processos i augmentant l'especificitat d'aquesta interacció. Aquesta modificació genètica de L. lactis ha estat àmpliament caracteritzada per provar les seves característiques com a interfase funcional vivent. S'ha demostrat que L. lactis és capaç d'expressar el fragment FNIII 7-10-GFP covalentment ancorat a la paret cel·lular bacteriana, havent-se caracteritzat també la seva activitat biològica amb tècniques com Western blot, ELISA i immunofluorescència. A més, aquest cep manté la capacitat de desenvolupament de biofilms, comunitats de bacteris sèssils adherits a un substrat que poden ser utilitzades com a interfase física entre cèl·lules de mamífer i substrats abiòtics. També s'ha estudiat la resposta cel·lular a la fibronectina expressada en la paret cel·lular de L. lactis. El estudi es va fer utilitzant diverses línies cel·lulars, com fibroblasts Fn-/Fn- i NIH3T3, mioblasts C2C12 i cèl·lules mesenquimals humanes derivades de medul·la òssia. Aquesta interfase vivent va ser capaç de provocar resposta cel·lular en forma d'adhesió a totes les línies estudiades, a més d'induir diferenciació de mioblasts a miotubs en C2C12 i de provocar la fosforilació de FAK, un marcador de senyalització cel·lular mediat per integrines, en les línies assajades. En cèl·lules mesenquimals humanes es va demostrar la capacitat del fragment de fibronectina exposat per fosforilar ERK1/2, una kinasa pertanyent a la ruta de senyalització MAPK, ruta que forma part de molts processos cel·lulars importants com diferenciació, proliferació i migració. Malgrat tot, aquesta tesi mostra només una prova de concepte d'un sistema que pot ser utilitzat per expressar gairebé qualsevol proteïna o molècula petita desitjada, que pot ser molt útil en el desenvolupament de nous teixits a partir de les seves cèl·lules progenitores. Aquestes molècules poden ser secretades en el medi o ancorades a la paret cel·lular, de manera constitutiva o sota demanda, a causa de la flexibilitat i àmplia varietat de sistemes d'expressió disponibles per L. lactis Aquesta biointerfase basada en bacteris vius estableix un nou paradigma en el camp de la funcionalització de superfícies per a aplicacions d'enginyería biomèdica.Rodrigo Navarro, A. (2015). Functional living biointerfaces to direct cell-material interaction [Tesis doctoral no publicada]. Universitat Politècnica de València. https://doi.org/10.4995/Thesis/10251/51461TESI

    Borax induces osteogenesis by stimulating NaBC1 transporter via activation of BMP pathway

    Get PDF
    [EN] The intrinsic properties of mesenchymal stem cells (MSCs) make them ideal candidates for tissue engineering applications. Efforts have been made to control MSC behavior by using material systems to engineer synthetic extracellular matrices and/or include soluble factors in the media. This work proposes a simple approach based on ion transporter stimulation to determine stem cell fate that avoids the use of growth factors. Addition of borax alone, transported by the NaBC1-transporter, enhanced MSC adhesion and contractility, promoted osteogenesis and inhibited adipogenesis. Stimulated-NaBC1 promoted osteogenesis via the BMP canonical pathway (comprising Smad1/YAP nucleus translocation and osteopontin expression) through a mechanism that involves simultaneous NaBC1/BMPR1A and NaBC1/alpha (5)beta (1)/alpha (v)beta (3) co-localization. We describe an original function for NaBC1 transporter, besides controlling borate homeostasis, capable of stimulating growth factor receptors and fibronectin-binding integrins. Our results open up new biomaterial engineering approaches for biomedical applications by a cost-effective strategy that avoids the use of soluble growth factors. Rico et al. propose a simple approach based on borax stimulation of NaBC1 transporter, which enhances FN-binding integrin-dependent mesenchymal stem cell adhesion and contractility, promotes osteogenesis and inhibits adipogenesis. Osteogenic differentiation depends on activation of the BMP pathway through a mechanism that involves simultaneous co-localization of NaBC1 with FN-binding integrins and BMPR1A.P.R. acknowledges support from the Spanish Ministry of Science, Innovation and Universities (RTI2018-096794), and Fondo Europeo de Desarrollo Regional (FEDER). CIBER-BBN is an initiative funded by the VI National R&D&I Plan 2008-2011, Iniciativa Ingenio 2010, Consolider Program, CIBER Actions and financed by the Instituto de Salud Carlos III with assistance from the European Regional Development Fund. M.S.S. acknowledges support from the UK Engineering and Physical Sciences Research Council (EPSRC-EP/P001114/1).Rico Tortosa, PM.; Rodrigo Navarro, A.; Sánchez-Pérez, L.; Salmerón Sánchez, M. (2020). Borax induces osteogenesis by stimulating NaBC1 transporter via activation of BMP pathway. Communications Biology. 3(1):1-15. https://doi.org/10.1038/s42003-020-01449-4S11531Akhurst, R. J. & Hata, A. Targeting the TGFbeta signalling pathway in disease. Nat. Rev. Drug Discov. 11, 790–811 (2012).Brizzi, M. F., Tarone, G. & Defilippi, P. Extracellular matrix, integrins, and growth factors as tailors of the stem cell niche. Curr. Opin. Cell Biol. 24, 645–651 (2012).Watt, F. M. & Huck, W. T. S. Role of extracellular matrix regulating stem cell fate. Nat. Rev. Mol. Cell Biol. 14, 467–473 (2013).Benoit, D. S. W., Schwartz, M. P., Durney, A. R. & Anseth, K. S. Small functional groups for controlled differentiation of hydrogel-encapsulated human mesenchymal stem cells. Nat. Mater. 7, 816–823 (2008).Baker, B. M., Trappmann, B., Wang, W. Y., Sakar, M. S., Kim, I. L., Shenoy, V. B., Burdick, J. A. & Chen, C. S. Cell-mediated fibre recruitment drives extracellular matrix mechanosensing in engineered fibrillary microenvironments. Nat. Mater. 14, 1262–1268 (2015).Das, R. K., Gocheva, V., Hammink, R., Zouani, O. F. & Rowan, A. E. Stress-stiffening-mediated stem-cell commitment switch in soft responsive hydrogels. Nat. Mater. 15, 318–325 (2015).Kilian, K. A., Bugarija, B., Lahn, B. T. & Mrksich, M. Geometric cues for directing the differentiation of mesenchymal stem cells. Proc. Natl Acad. Sci. USA 107, 4872–4877 (2010).Yang, J., McNamara, L. E., Gadegaard, N., Alakpa, E. V., Burgess, K. V., Dominic Meek, R. M. & Dalby, M. J. Nanotopographical induction of osteogenesis through adhesion, bone morphogenetic protein cosignaling, and regulation of microRNAs. ACS Nano. 8, 9941–9953 (2014).Dalby, M. J., García, A. J. & Salmeron-Sanchez, M. Receptor control in mesenchymal stem cell engineering. Nat. Rev. 3, 17091 (2018).Carragee, E. J., Hurwitz, E. L. & Weiner, B. K. A critical review of recombinant human bone morphogenetic protein-2 trials in spinal surgery: emerging safety concerns and lessons learned. Spine J. 11, 471–491 (2011).Vining, K. H. & Mooney, D. J. Mechanical forces direct stem cell behavior in development and regeneration. Nat. Rev. 18, 728–742 (2017).Biggs, M. J., Richards, R. G., Gadegaard, N., Wilkinson, C. D., Oreffo, R. O. & Dalby, M. J. The use of nanoscale topography to modulate the dynamics of adhesion formation in primary osteoblasts and ERK/MAPK signaling in STRO-1+ enriched skeletal stem cells. Biomaterials 30, 5094–5103 (2009).McBeath, R., Pirone, D. M., Nelson, C. M., Bhadriraju, K. & Chen, C. S. Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev. Cell. 6, 483–495 (2004).Hille, B. Ion Channels of Excitable Membranes. (Sinauer Associates Inc, Sunderland, MA, 2001).Lauritzen, I., Chemin, J., Honoré, E., Martine, J., Guy, N., Lazdunski, M. & Patel, A. J. Cross-talk between the mechano-gated K2p channel TREK-1 and the actin cytoskeleton. EMBO Rep. 6, 642–648 (2005).Gasparski, A. N. & Beningo, K. A. Mechanoreception at the cell membrane: more than the integrins. Arch. Biochem. Biophys. 586, 20–26 (2015).Pillozzi, S. & Becchetti, A. Ion channels in hematopoietic and mesenchymal stem cells. Stem Cells Int. 2012, 217910 (2012).Park, M., Li, Q., Shcheynikov, N., Zeng, W. & Muallem, S. NaBC1 is a ubiquitous electrogenic Na+-coupled borate transporter essential for cellular boron homeostasis and cell growth and proliferation. Mol. Cell. 16, 331–341 (2004).Vithana, E. N., Morgan, P., Sundaresan, P., Ebenezer, N. D., Tan, D. T., Mohamed, M. D., Anand, S., Khine, K. O., Venkataraman, D., Yong, V. H., Salto-Tellez, M., Venkatraman, A., Guo, K., Hemadevi, B., Srinivasan, M., Prajna, V., Khine, M., Casey, J. R., Inglehearn, C. F. & Aung, T. Mutations in sodium-borate cotransporter SLC4A11 cause recessive congenital hereditary endotelial dystrophy (CHED2). Nat. Genet. 38, 755–757 (2006).Lopez, I. A., Rosenblatt, M. I., Kim, C., Galbraith, G. C., Jones, S. M., Kao, L., Newman, D., Liu, W., Yeh, S., Pushkin, A., Abuladze, N. & Kurtz, I. Slc4a11 gene disruption in mice: cellular targets of sensorineural abnormalities. J. Biol. Chem. 284, 26882–26896 (2009).Rico, P., Rodrigo-Navarro, A. & Salmeron-Sanchez, M. Borax-loaded PLLA for promotion of myogenic differentiation. Tissue Eng. Part A. 21, 2662–2672 (2015).Rico, P., Rodrigo-Navarro, A., de la Peña, M., Moulisová, V., Costell, M. & Salmeron-Sanchez, M. Simultaneous boron ion-channel activation for enhanced vascularization. Adv. Biosyst. 3, 1800220 (2019).Cifti, E., Köse, S., Korkusuz, P., Timuçin, M. & Korkusuz, F. Boron containing nano hydroxyapatites (Bn-HAp) stimulate mesenchymal stem cell adhesion, proliferation and differentiation. Key Eng. Mater. 631, 373–378 (2015).Li, X., Wang, X., Jiang, X., Yamaguchi, M., Ito, A., Bando, Y. & Golberg, D. Boron nitride nanotube-enhanced osteogenic differentiation of mesenchymal stem cells. J. Biomed. Res. 104, 323–329 (2015).Liu, Y. J., Su, W. T. & Chen, P. H. Magnesium and zinc borate enhance osteoblastic differentiation of stem cells from human exfoliated deciduous teeth in vitro. J. Biomater. Appl. 32, 765–774 (2018).Dogan, A., Demirci, S., Apdik, H., Bayrak, O. F., Gulluoglu, S., Tuysuz, E. C., Gusev, O., Rizanov, A. A., Nikerel, E. & Sahin, F. A new hope for obesity management: boron inhibits adipogenesis in progenitor cells through the Wnt/β-catenin pathway. Metabolism 69, 130–142 (2017).Abdik, E. A., Abdik, H., Tasli, P. N., Asli, A., Deniz, H. & Sahin, F. Suppressive role of boron on adipogenic differentiation and fat deposition in human mesenchymal stem cells. Biol. Trace Elem. Res. 188, 384–392 (2018).Humphries, M. J., Travis, M. A., Clark, K. & Mould, A. P. Mechanisms of integration of cells and extracellular matrices by integrins. Biochem. Soc. Trans. 32, 822–825 (2004).Burns, A. E. & Varin, J. Poly-L-lactic acid rod fixation results in foot surgery. J. Foot Ankle Surg. 37, 37–41 (1998).Harada, S. & Rodan, G. A. Control of osteoblast function and regulation of bone mass. Nature 423, 349–355 (2003).Gregory, C. A., Ylostalo, J. & Prockop, D. J. Adult bone marrow stem/progenitor cells (MSCs) are preconditioned by microenvironmental niches in culture: a two-stage hypothesis for regulation of MSC fate. Sci. Stke. 294, pe37 (2005).Jones, D. R. H. & Ashby, M. F. Engineering Materials 1. (Butterworth-Heinemann, 2019).Farah, S., Anderson, D. G. & Langer, R. Physical and mechanical properties of PLA, and their functions in widespread applications-A comprehensive review. Adv. Drug Deliv. Rev. 107, 367–392 (2016).González-García, C., Moratal, D., Oreffo, R. O. C., Dalby, M. J. & Salmeron-Sanchez, M. Surface mobility regulates skeletal stem cell differentiation. Integr. Biol. 4, 531–539 (2012).Liddington, R. C. & Ginsberg, M. H. Integrin activation takes shape. J. Cell Biol. 158, 833–839 (2002).Ganor, Y., Besser, M. & Ben-Zakay, N. et al. Human T cells express a functional ionotropic glutamate receptor GluR3, and glutamate by itself triggers integrin-mediated adhesion to laminin and fibronectin and chemotactic migration. J. Immunol. 170, 4362–4372 (2003).Puklin-Faucher, E. & Sheetz, M. P. The mechanical integrin cycle. J. Cell Sci. 122, 179–186 (2009).Liao, S. F., Monegue, J. S., Lindemann, M. D., Cromwell, G. L. & Matthews, J. C. Dietary supplementation of boron differentially alters expression of borate transporter (NaBC1) mRNA by jejunum and kidney of growing pigs. Biol. Trace Elem. Res. 143, 901–912 (2011).Saidak, Z., Le Henaff, C., Azzi, S., Marty, C., Da Nascimento, S., Sonnet, P. & Marie, P. J. Wnt/β-catenin signaling mediates osteoblast differentiation triggered by peptide-induced α5β1 integrin priming in Mesenchymal Skeletal Cells. J. Biol. Chem. 290, 6903–6912 (2015).Chen, Q., Shou, P., Zhang, L., Xu, C., Zheng, C., Han, Y., Li, W., Huang, Y., Zhang, X., Shao, C., Roberts, A. I., Rabson, A. B., Ren, G., Zhang, Y., Wang, Y., Denhardt, D. T. & Shi, Y. An osteopontine-integrin interaction plays a critical role in directing adipogenesis and osteogenesis by mesenchymal stem cells. Stem Cells 32, 327–337 (2014).Hofmann, G., Bernabei, P. A. & Crociani, O. et al. HERG K+ channels activation during β1 integrin-mediated adhesion to fibronectin induces an up-regulation of αvβ3 integrin in the preosteoclastic leukemia cell line FLG 29.1. J. Biol. Chem. 276, 4923–4931 (2001).Becchetti, A. et al. Response to fibronectin-integrin interaction in leukaemia cells: delayed enhancing of a K + current. Proc. R. Soc. Lond. 248, 235–240 (1992).Arcangeli, A. & Becchetti, A. Complex functional interaction between integrin receptors and ion channels. TRENDS Cell Biol. 16, 631–639 (2006).Jing, J., Hinton, R. J. & Feng, J. Q. BMR1A signaling in cartilage development and endochondral bone formation. Vitam. Hormones. 99, 273–291 (2015).Kimura, M., Ito, M., Amano, K., Chihara, Y., Fukata, M., Nakafuku, B., Yamamori, J., Feng, J., Nakano, T. & Okawa, K. et al. Regulation of myosin phosphatase by Rho and Rho-associated kinase (Rho-kinase). Science 273, 245–248 (1996).Pelham, R. J. & Wang, Y. Cell locomotion and focal adhesions are regulated by substrate flexibility. Proc. Natl Acad. Sci. USA 94, 13661–13665 (1997).Kovacs, M., Tóth, J., Hetényi, C., Málnási-Csizmadia, A. & Sellers, J. Mechanism of blebbistatin inhibition of myosin II. J. Biol. Chem. 279, 35557–35563 (2004).Narumiya, S., Ishizaki, T. & Ufhata, M. Use and properties of ROCK-specific inhibitor Y-27632. Methods Enzymol. 325, 273–284 (2000).Schmierer, B. & Hill, C. S. TGFbeta-SMAD signal transduction: molecular specificity and functional flexibility. Nat. Rev. 8, 970–982 (2007).Zhao, B., Li, L. & Guan, K. L. Hippo signaling at a glance. J. Cell Sci. 123, 4001–4006 (2010).Varelas, X. The Hippo pathway effectors TAZ and YAP in development, homeostasis and disease. Development 141, 1614–1626 (2014).Pittenger, M. F., Mackay, A. M., Beck, S. C., Jaiswal, R. K., Douglas, R., Mosca, J. D., Moorman, M. A., Simonetti, D. W., Craig, S. & Marshak, D. R. Multilineage potential of adult human mesenchymal stem cells. Science 284, 143–147 (1999).Kirkham, G. R. & Cartmell, S. H. Genes and proteins involved in the regulation of osteogenesis. Top. Tissue Eng. 3, 1–22 (2007).Chamberlain, G., Fox, J., Ashton, B. & Middleton, J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells 25, 2739–2749 (2007).Lowe, C. E., O´Rahilly, S. & Rochford, J. J. Adipogenesis at a glance. J. Cell Sci. 124, 2681–2686 (2011).MacQueen, L., Sun, Y. & Simmons, C. A. Mesenchymal stem cell mechanobiology and emerging experimental platforms. J. R. Soc. Interface 10, 20130179 (2013).Ivanovska, I. L., Shin, J. W., Swift, J. & Discher, D. E. Stem cell mechanobiology: diverse lessons from bone marrow. Trends Cell Biol. 25, 523–532 (2015).Phimphilai, M., Zhao, Z., Boules, H., Roca, H. & Franceschi, R. T. BMP signaling is required for RUNX2-dependent induction of the osteoblast phenotype. J. Bone Miner. Res. 21, 637–646 (2006).Comoglio, P. M., Boccaccio, C. & Trusolino, L. Interactions between growth factor receptors and adhesion molecules: breaking the rules. Curr. Opin. Cell Biol. 15, 565–571 (2003).Fourel, L., Valat, A., Faurobert, E., Guillot, R., Bourrin-Reynard, I., Ren, K., Lafanechère, L., Planus, E., Picart, C. & Albiges-Rizo, C. β3 integrin-mediated spreading induced by matrix-bound BMP-2 controls Smad signaling in a stiffness-independent manner. J. Cell Biol. 212, 693–706 (2016).Morandi, E. M., Verstappen, R., Zwierzina, M. E., Geley, S., Pierer, G. & Ploner, C. ITGAV and ITGA5 diversely regulate proliferation and adipogenic differentiation of human adipose derived stem cells. Sci. Rep. 6, 28889 (2016).Brazil, D. P., Church, R. H., Surae, S., Godson, C. & Martin, F. BMP signalling: agony and antagony in the family. Trends Cell Biol. 25, 249–264 (2015).Nardone, G., Oliver-De La Cruz, J., Vrbsky, J., Martini, C., Pribyl, J., Skla´dal, P., Pesl, M., Caluori, G., Pagliari, S., martino, F., Maceckova, Z., Hajduch, M., Sanz-Garcia, A., Pugno, N. M., Stokin, G. B. & Forte, G. YAP regulates cell mechanics by controlling focal adhesion assembly. Nat. Commun. 8, 15321 (2017).Miyazono, K., Maeda, S. & Imamura, T. BMP receptor signaling: transcriptional targets, regulation of signals, and signaling cross-talk. Cytokine Growth Factor Rev. 16, 251–263 (2005).Rico, P., Rodrigo-Navarro, A., Sánchez Pérez, L. & Salmeron-Sanchez, M. Borax induces osteogenesis by stimulating NaBC1 transporter via activation of BMP pathway. Commun. Biol. https://doi.org/10.5525/gla.researchdata.1076 (2020)

    Engineered microenvironments for synergistic VEGF - integrin signalling during vascularization

    Get PDF
    We have engineered polymer-based microenvironments that promote vasculogenesis both in vitro and in vivo through synergistic integrin-growth factor receptor signalling. Poly(ethyl acrylate) (PEA) triggers spontaneous organization of fibronectin (FN) into nanonetworks which provide availability of critical binding domains. Importantly, the growth factor binding (FNIII12-14) and integrin binding (FNIII9-10) regions are simultaneously available on FN fibrils assembled on PEA. This material platform promotes synergistic integrin/VEGF signalling which is highly effective for vascularization events in vitro with low concentrations of VEGF. VEGF specifically binds to FN fibrils on PEA compared to control polymers (poly(methyl acrylate), PMA) where FN remains in a globular conformation and integrin/GF binding domains are not simultaneously available. The vasculogenic response of human endothelial cells seeded on these synergistic interfaces (VEGF bound to FN assembled on PEA) was significantly improved compared to soluble administration of VEGF at higher doses. Early onset of VEGF signalling (PLCγ1 phosphorylation) and both integrin and VEGF signalling (ERK1/2 phosphorylation) were increased only when VEGF was bound to FN nanonetworks on PEA, while soluble VEGF did not influence early signalling. Experiments with mutant FN molecules with impaired integrin binding site (FN-RGE) confirmed the role of the integrin binding site of FN on the vasculogenic response via combined integrin/VEGF signalling. In vivo experiments using 3D scaffolds coated with FN and VEGF implanted in the murine fat pad demonstrated pro-vascularization signalling by enhanced formation of new tissue inside scaffold pores. PEA-driven organization of FN promotes efficient presentation of VEGF to promote vascularization in regenerative medicine applications

    Simultaneous boron ion-channel/growth factor receptor activation for enhanced vascularization

    Get PDF
    [EN] Boron ion is essential in metabolism and its concentration is regulated by ion-channel NaBC1. NaBC1 mutations cause corneal dystrophies such as Harboyan syndrome. Here we propose a 3D molecular model for NaBC1 and show that simultaneous stimulation of NaBC1 and vascular growth factor receptors (VEGFR) promote angiogenesis in vitro and in vivo with ultra-low concentrations of VEGF. We show Human Umbilical Vein Endothelial Cells (HUVEC) organization into tubular structures indicative of vascularization potential. Enhanced cell sprouting was found only in the presence of VEGF and boron, effect abrogated after blocking NaBC1. We demonstrate that stimulated NaBC1 promotes angiogenesis via PI3k-independent pathways and that ¿5ß1/¿vß3-integrin binding is not essential to enhanced HUVEC organization. We describe a novel vascularization mechanism that involves the crosstalk and colocalization between NaBC1/VEGFR receptors. This has important translational consequences: just by administering boron, taking advantage of endogenous VEGF, in vivo vascularization is shown in a chorioallantoic membrane assay.P.R. acknowledges support from the Ministerio de Economia, Industria y Competitividad, Gobierno de Espana (MINECO) (MAT2015-69315-C3-1-R), and European Regional Development Fund (FEDER). CIBER-BBN is an initiative funded by the VI National R&D&I Plan 2008-2011, Iniciativa Ingenio 2010, Consolider Program, CIBER Actions and financed by the Instituto de Salud Carlos III with assistance from the European Regional Development Fund. M. S. S. acknowledges support from the European Research Council (ERC-HealInSynergy 306990) and the UK Engineering and Physical Sciences Research Council (EPSRC-EP/P001114/1). The authors are very grateful to Productos Florida farm for kindly providing chick embryos for CAM assay.Rico Tortosa, PM.; Rodrigo Navarro, A.; La Peña Del Rivero, MD.; Moulisova, V.; Costell, M.; Salmerón Sánchez, M. (2018). Simultaneous boron ion-channel/growth factor receptor activation for enhanced vascularization. Advanced Biosystems. 3(1):1-12. https://doi.org/10.1002/adbi.201800220S11231Yancopoulos, G. D., Davis, S., Gale, N. W., Rudge, J. S., Wiegand, S. J., & Holash, J. (2000). Vascular-specific growth factors and blood vessel formation. Nature, 407(6801), 242-248. doi:10.1038/35025215Carmeliet, P. (2005). Angiogenesis in life, disease and medicine. Nature, 438(7070), 932-936. doi:10.1038/nature04478Moulisová, V., Gonzalez-García, C., Cantini, M., Rodrigo-Navarro, A., Weaver, J., Costell, M., … Salmerón-Sánchez, M. (2017). Engineered microenvironments for synergistic VEGF – Integrin signalling during vascularization. Biomaterials, 126, 61-74. doi:10.1016/j.biomaterials.2017.02.024Briquez, P. S., Clegg, L. E., Martino, M. M., Gabhann, F. M., & Hubbell, J. A. (2016). Design principles for therapeutic angiogenic materials. Nature Reviews Materials, 1(1). doi:10.1038/natrevmats.2015.6Hanft, J. R., Pollak, R. A., Barbul, A., Gils, C. va., Kwon, P. S., Gray, S. M., … Breen, T. J. (2008). Phase I trial on the safety of topical rhVEGF on chronic neuropathic diabetic foot ulcers. Journal of Wound Care, 17(1), 30-37. doi:10.12968/jowc.2008.17.1.27917Woo, E. J. (2012). Recombinant human bone morphogenetic protein-2: adverse events reported to the Manufacturer and User Facility Device Experience database. The Spine Journal, 12(10), 894-899. doi:10.1016/j.spinee.2012.09.052United States Food and Drug Administration Product Description Regranex https://www.fda.gov/downloads/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/UCM142821.avi (accessed: May2008).Carmeliet, P., & Jain, R. K. (2011). Molecular mechanisms and clinical applications of angiogenesis. Nature, 473(7347), 298-307. doi:10.1038/nature10144Hynes, R. O. (2002). Integrins. Cell, 110(6), 673-687. doi:10.1016/s0092-8674(02)00971-6Mahabeleshwar, G. H., Feng, W., Reddy, K., Plow, E. F., & Byzova, T. V. (2007). Mechanisms of Integrin–Vascular Endothelial Growth Factor Receptor Cross-Activation in Angiogenesis. Circulation Research, 101(6), 570-580. doi:10.1161/circresaha.107.155655Olsson, A.-K., Dimberg, A., Kreuger, J., & Claesson-Welsh, L. (2006). VEGF receptor signalling ? in control of vascular function. Nature Reviews Molecular Cell Biology, 7(5), 359-371. doi:10.1038/nrm1911Alexander, R. A., Prager, G. W., Mihaly-Bison, J., Uhrin, P., Sunzenauer, S., Binder, B. R., … Breuss, J. M. (2012). VEGF-induced endothelial cell migration requires urokinase receptor (uPAR)-dependent integrin redistribution. Cardiovascular Research, 94(1), 125-135. doi:10.1093/cvr/cvs017Herkenne, S., Paques, C., Nivelles, O., Lion, M., Bajou, K., Pollenus, T., … Struman, I. (2015). The interaction of uPAR with VEGFR2 promotes VEGF-induced angiogenesis. Science Signaling, 8(403), ra117-ra117. doi:10.1126/scisignal.aaa2403Lauritzen, I., Chemin, J., Honoré, E., Jodar, M., Guy, N., Lazdunski, M., & Jane Patel, A. (2005). Cross‐talk between the mechano‐gated K 2P channel TREK‐1 and the actin cytoskeleton. EMBO reports, 6(7), 642-648. doi:10.1038/sj.embor.7400449Gasparski, A. N., & Beningo, K. A. (2015). Mechanoreception at the cell membrane: More than the integrins. Archives of Biochemistry and Biophysics, 586, 20-26. doi:10.1016/j.abb.2015.07.017Munaron, L., Genova, T., Avanzato, D., Antoniotti, S., & Fiorio Pla, A. (2012). Targeting Calcium Channels to Block Tumor Vascularization. Recent Patents on Anti-Cancer Drug Discovery, 8(1), 27-37. doi:10.2174/1574892811308010027Yao, X., & Garland, C. J. (2005). Recent Developments in Vascular Endothelial Cell Transient Receptor Potential Channels. Circulation Research, 97(9), 853-863. doi:10.1161/01.res.0000187473.85419.3eRico, P., Rodrigo-Navarro, A., & Salmerón-Sánchez, M. (2015). Borax-Loaded PLLA for Promotion of Myogenic Differentiation. Tissue Engineering Part A, 21(21-22), 2662-2672. doi:10.1089/ten.tea.2015.0044Park, M., Li, Q., Shcheynikov, N., Zeng, W., & Muallem, S. (2004). NaBC1 Is a Ubiquitous Electrogenic Na+-Coupled Borate Transporter Essential for Cellular Boron Homeostasis and Cell Growth and Proliferation. Molecular Cell, 16(3), 331-341. doi:10.1016/j.molcel.2004.09.030Vithana, E. N., Morgan, P., Sundaresan, P., Ebenezer, N. D., Tan, D. T. H., Mohamed, M. D., … Aung, T. (2006). Mutations in sodium-borate cotransporter SLC4A11 cause recessive congenital hereditary endothelial dystrophy (CHED2). Nature Genetics, 38(7), 755-757. doi:10.1038/ng1824Lopez, I. A., Rosenblatt, M. I., Kim, C., Galbraith, G. C., Jones, S. M., Kao, L., … Kurtz, I. (2009). Slc4a11Gene Disruption in Mice. Journal of Biological Chemistry, 284(39), 26882-26896. doi:10.1074/jbc.m109.008102Parker, M. D., Ourmozdi, E. P., & Tanner, M. J. A. (2001). Human BTR1, a New Bicarbonate Transporter Superfamily Member and Human AE4 from Kidney. Biochemical and Biophysical Research Communications, 282(5), 1103-1109. doi:10.1006/bbrc.2001.4692Zangi, R., & Filella, M. (2012). Transport routes of metalloids into and out of the cell: A review of the current knowledge. Chemico-Biological Interactions, 197(1), 47-57. doi:10.1016/j.cbi.2012.02.001Tanjore, H., Zeisberg, E. M., Gerami-Naini, B., & Kalluri, R. (2007). β1 integrin expression on endothelial cells is required for angiogenesis but not for vasculogenesis. Developmental Dynamics, 237(1), 75-82. doi:10.1002/dvdy.21385Gerber, H.-P., Dixit, V., & Ferrara, N. (1998). Vascular Endothelial Growth Factor Induces Expression of the Antiapoptotic Proteins Bcl-2 and A1 in Vascular Endothelial Cells. Journal of Biological Chemistry, 273(21), 13313-13316. doi:10.1074/jbc.273.21.13313Tan, C., Cruet-Hennequart, S., Troussard, A., Fazli, L., Costello, P., Sutton, K., … Dedhar, S. (2004). Regulation of tumor angiogenesis by integrin-linked kinase (ILK). Cancer Cell, 5(1), 79-90. doi:10.1016/s1535-6108(03)00281-2George, E. L., Baldwin, H. S., & Hynes, R. O. (1997). Fibronectins Are Essential for Heart and Blood Vessel Morphogenesis But Are Dispensable for Initial Specification of Precursor Cells. Blood, 90(8), 3073-3081. doi:10.1182/blood.v90.8.3073Fassler, R., & Meyer, M. (1995). Consequences of lack of beta 1 integrin gene expression in mice. Genes & Development, 9(15), 1896-1908. doi:10.1101/gad.9.15.1896Soldi, R., Mitola, S., Strasly, M., Defilippi, P., Tarone, G., & Bussolino, F. (1999). Role of αvβ3 integrin in the activation of vascular endothelial growth factor receptor-2. The EMBO Journal, 18(4), 882-892. doi:10.1093/emboj/18.4.882Takahashi, S., Leiss, M., Moser, M., Ohashi, T., Kitao, T., Heckmann, D., … Fässler, R. (2007). The RGD motif in fibronectin is essential for development but dispensable for fibril assembly. Journal of Cell Biology, 178(1), 167-178. doi:10.1083/jcb.200703021Ribatti, D. (2008). Chapter 5 Chick Embryo Chorioallantoic Membrane as a Useful Tool to Study Angiogenesis. International Review of Cell and Molecular Biology, 181-224. doi:10.1016/s1937-6448(08)01405-6Novosel, E. C., Kleinhans, C., & Kluger, P. J. (2011). Vascularization is the key challenge in tissue engineering. Advanced Drug Delivery Reviews, 63(4-5), 300-311. doi:10.1016/j.addr.2011.03.004García, J. R., & García, A. J. (2015). Biomaterial-mediated strategies targeting vascularization for bone repair. Drug Delivery and Translational Research, 6(2), 77-95. doi:10.1007/s13346-015-0236-0Briquez, P. S., Hubbell, J. A., & Martino, M. M. (2015). Extracellular Matrix-Inspired Growth Factor Delivery Systems for Skin Wound Healing. Advances in Wound Care, 4(8), 479-489. doi:10.1089/wound.2014.0603Simón-Yarza, T., Formiga, F. R., Tamayo, E., Pelacho, B., Prosper, F., & Blanco-Prieto, M. J. (2012). Vascular Endothelial Growth Factor-Delivery Systems for Cardiac Repair: An Overview. Theranostics, 2(6), 541-552. doi:10.7150/thno.3682Kargozar, S., Baino, F., Hamzehlou, S., Hill, R. G., & Mozafari, M. (2018). Bioactive Glasses: Sprouting Angiogenesis in Tissue Engineering. Trends in Biotechnology, 36(4), 430-444. doi:10.1016/j.tibtech.2017.12.003Laplante, M., & Sabatini, D. M. (2009). mTOR signaling at a glance. Journal of Cell Science, 122(20), 3589-3594. doi:10.1242/jcs.051011Byzova, T. V., Goldman, C. K., Pampori, N., Thomas, K. A., Bett, A., Shattil, S. J., & Plow, E. F. (2000). A Mechanism for Modulation of Cellular Responses to VEGF. Molecular Cell, 6(4), 851-860. doi:10.1016/s1097-2765(05)00076-

    Simultaneous boron ion-channel/growth factor receptor activation for enhanced vascularization

    Get PDF
    Boron ion is essential in metabolism and its concentration is regulated by ion-channel NaBC1. NaBC1 mutations cause corneal dystrophies such as Harboyan syndrome. Here we propose a 3D molecular model for NaBC1 and show that simultaneous stimulation of NaBC1 and vascular growth factor receptors (VEGFR) promote angiogenesis in vitro and in vivo with ultra-low concentrations of VEGF. We show Human Umbilical Vein Endothelial Cells (HUVEC) organization into tubular structures indicative of vascularization potential. Enhanced cell sprouting was found only in the presence of VEGF and boron, effect abrogated after blocking NaBC1. We demonstrate that stimulated NaBC1 promotes angiogenesis via Akt-independent pathways and that α5β1/αvβ3-integrin binding is not essential to enhanced HUVEC organization. We describe a novel vascularization mechanism that involves the crosstalk and colocalization between NaBC1/VEGFR receptors. This has important translational consequences: just by administering boron, taking advantage of endogenous VEGF, in vivo vascularization is shown in a chorioallantoic membrane assay

    A hydrogel platform that incorporates laminin isoforms for efficient presentation of growth factors – neural growth and osteogenesis

    Get PDF
    Laminins (LMs) are important structural proteins of the extracellular matrix (ECM). The abundance of every LM isoform is tissue‐dependent, suggesting that LM has tissue‐specific roles. LM binds growth factors (GFs), which are powerful cytokines widely used in tissue engineering due to their ability to control stem cell differentiation. Currently, the most commonly used ECM mimetic material in vitro is Matrigel, a matrix of undefined composition containing LM and various GFs, but subjected to batch variability and lacking control of physicochemical properties. Inspired by Matrigel, a new and completely defined hydrogel platform based on hybrid LM‐poly(ethylene glycol) (PEG) hydrogels with controllable stiffness (1–25 kPa) and degradability is proposed. Different LM isoforms are used to bind and efficiently display GFs (here, bone morphogenetic protein (BMP‐2) and beta‐nerve growth factor (β‐NGF)), enabling their solid‐phase presentation at ultralow doses to specifically target a range of tissues. The potential of this platform to trigger stem cell differentiation toward osteogenic lineages and stimulate neural cells growth in 3D, is demonstrated. These hydrogels enable 3D, synthetic, defined composition, and reproducible cell culture microenvironments reflecting the complexity of the native ECM, where GFs in combination with LM isoforms yield the full diversity of cellular processes

    Living biointerfaces for the maintenance of mesenchymal stem cell phenotypes

    Get PDF
    Living interfaces are established as a novel class of active materials that aim to provide an alternative to traditional static cell culture methods by enabling users to accurately control cell behaviour in a precise, dynamic, and reliable system-internal manner. To this day, the only reported biointerface has been a coculture between a biofilm of nonpathogenic genetically engineered bacteria and mammalian cells, where the recombinant proteins produced by the bacteria directly influence cell behaviour. In this work, a biointerface is presented between Lactococcus lactis (L. lactis) and human mesenchymal stem cells (hMSCs). L. lactis have been engineered to produce human C-X-C motif chemokine ligand 12, thrombopoietin, vascular cell adhesion protein 1, and the 7th–10th type III domains of human fibronectin, with the aim of recreating the native bone marrow conditions ex vivo. This active microenvironment has been shown to maintain key hMSC stemness markers, preventing their osteogenic and adipogenic differentiation, and maintaining high stem cell viability and physiological cell-to-substrate adhesion dynamics. This work presents proof of concept data that hMSC stemness can be regulated by living materials, using a system based on the symbiotic interaction between different engineered bacteria and mammalian cells

    Living biomaterials to engineer haematopoietic stem cell niches

    No full text
    Living biointerfaces are a new class of biomaterials combining living cells and polymeric matrices that can act as biologically active and instructive materials that host and provide signals to surrounding cells. Here we introduce living biomaterials based on Lactococcus lactis to control hematopoietic stem cells in 2D surfaces and 3D hydrogels. L. lactis has been modified to express C-X-C motif chemokine ligand 12 (CXCL12), thrombopoietin (TPO), vascular cell adhesion protein 1 (VCAM1) and the 7th-10th type III domains of human plasma fibronectin (FN III7-10), in an attempt to mimic ex vivo the conditions of the human bone marrow Our results suggest that living biomaterials that incorporate bacteria expressing recombinant CXCL12, TPO, VCAM1 and FN in both 2D systems direct hematopoietic stem and progenitor cells (HSPC)-bacteria interaction, and in 3D using hydrogels functionalized with full-length human plasma fibronectin allow for a notable expansion of the CD34+/CD38–/CD90+ HSPC population compared to the initial population. These results provide a strong evidence based on data that suggests the possibility of using living materials based on genetically engineered bacteria for the ex-vivo expansion of HSPC with eventual practical clinical applications in HSPC transplantation for hematological disorders
    corecore