• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 148
  • 34
  • 30
  • 15
  • 12
  • 6
  • 5
  • 5
  • 3
  • 2
  • 1
  • Tagged with
  • 347
  • 112
  • 63
  • 46
  • 46
  • 39
  • 36
  • 35
  • 35
  • 32
  • 29
  • 26
  • 26
  • 25
  • 24
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
261

Caractérisation cellulaire et moléculaire de l'activité de dérivés de 2-aryl-3-quinolone, une famille de petites molécules antagonistes de la queue cytoplasmique des intégrines / Cellular and molecular characterization of 2-aryl-3-quinolones derivatives, a small molecule family antagonist of integrin cytoplasmic tail

Fiorucci, Sandrine 16 September 2013 (has links)
Les flavonoïdes sont étudiés depuis des années pour leurs propriétés préventives et leur potentiel comme agents thérapeutiques. Plusieurs mécanismes pourraient intervenir dans leur activité anti-cancéreuse dont une inhibition de l'adhérence et de l'étalement cellulaire et une inhibition des propriétés invasives des cellules cancéreuses. Les dérivés de 3-aryl-2-quinolones sont structurellement proches des flavonoïdes et ont été caractérisés comme étant des composés anti-migratoires (Joseph et Al., J.Med.Chem, 2002). Comme la migration cellulaire est hautement dépendante des structures adhésives assemblées par la cellule, nous avons étudié l'activité de ces composés sur les adhérences focales et fibrillaires. Ces larges complexes protéiques impliquent notamment les intégrines et leurs partenaires cytoplasmiques les liant au cytosquelette. Les intégrines permettent à la cellule de percevoir son microenvironnement et de s'y adapter. Les structures d'adhérence contenant les intégrines sont en retour capables de contrôler cet environnement (dégradation matricielle, fibrillogenèse…). Nos travaux montrent que les dérivés de 3-aryl-2-quinolone sont capables d'inhiber l'étalement cellulaire et de provoquer le désassemblage de structures adhésives préalablement établies de façon dose-dépendante et indépendamment de la composition de la matrice extracellulaire. L'activité des composés est finement liée à leur structure et de légères modifications de la composition chimique de leur chaîne latérale peuvent inhiber leur activité. Nous avons pu établir une relation structure-activité pour cette famille de composés et avons étudié les mécanismes moléculaires menant au désassemblage des structures d'adhérences quand les cellules sont traitées par ces molécules. Des études par RMN ont montré une interaction directe entre le chef de file de cette famille de composés et le domaine cytoplasmique des intégrines à chaîne béta 3 et nous avons pu montrer que cette interaction inhibait la liaison sur l'intégrine de l'un de ces activateurs, la kindline. L'agrégation plaquettaire est dépendante de l'activation des intégrines et constitue un excellent système d'étude physiologique pour les inhibiteurs de ces récepteurs. Le chef de file des dérivés de 3-aryl-2-quinolone est capable d'inhiber l'agrégation plaquettaire et la formation du thrombus, ce qui en fait un bon candidat médicament comme anti-thrombotique. / Flavonoïds have been studied for years for their potential chemopreventive and chemotherapeutic action. Several mechanisms might account for their anticancer activity, among which inhibition of cell adhesion and spreading, or inhibition of tumor cell invasion. 3-aryl-2-quinolone derivatives are chemical structures close to flavonoïds and were first designed as anti-migratory agents (Joseph et Al., J.Med.Chem, 2002). As cell migration is highly dependent on the cell adhesion machinery, we decided to investigate the action of these molecules on focal and fibrillar adhesions. These large protein complexes include heterodimeric transmembrane proteins, the integrins, and their cytoplasmic interactors able to link to the cytoskeleton. Integrins allow microenvironment sensing and cellular response to it. Adhesive structures containing integrins are also able to control cell microenvironment (matrix degradation, fibrillogenesis…). Our studies show that 3-aryl-2-quinolone derivatives are able not only to prevent cell spreading but also to disrupt already well-established focal adhesions in a reversible and ECM composition independent manner. The activity of the molecule is closely linked with its structure, as very slight modification of the lateral chain of the compound can totally impair its activity. Our work is focused on establishing a Structure-Activity Relationship of 3-aryl-2-quinolone derivatives and on investigating the molecular mechanisms underlying this activity. Osteoblasts treatment by 3-aryl-2-quinolone derivatives triggers a rapid disassembly of focal and fibrillar adhesions. NMR experiments show a direct interaction between the lead compound of the family and 3 integrin cytoplasmic tail and pull-down assay show that it is able to reduce the interaction between 3 integrin and kindlin, one of its coactivator. As platelet activation is an archetype of 3 integrin activation, we tested the activity of 3-aryl-2-quinolone on this physiological process. Under treatment, platelets failed to become activated and are unable to trigger thrombus formation, providing an interest to the 3-aryl-2-quinolone derivatives as potential anti-thrombotic agents.
262

Diferentes respostas à alternagina-c, uma proteína tipo desintegrina, em fibroblastos, células tumorais de mama e células endoteliais in vitro

Santos, Lívia Mara 10 December 2013 (has links)
Made available in DSpace on 2016-06-02T19:22:10Z (GMT). No. of bitstreams: 1 5684.pdf: 2471155 bytes, checksum: f035dbceafb88325d3cb0d3fb8280603 (MD5) Previous issue date: 2013-12-10 / Financiadora de Estudos e Projetos / Matrix metalloproteinases (MMPs) are key factors in tumor progression that allow tumor cells to modify the extracellular matrix (ECM) and to release cytokines, growth factors being activated by cell surface molecules such as the integrins. Integrins are major adhesion receptors of cell surface that connect the cells to the external environment enabling its movement. Integrins activate signaling cascades that influence the adhesion, survival and cell proliferation. Important inhibitors of these molecules were found in snake venoms called disintegrins. Alternagin-C (ALT-C) a disintegrin from Rhinocerophis alternatus snake venom has affinity with α2β1 integrin therfore modulating cell adhesion, migration and proliferation. However, the effect of ALT-C on MMP activity has not been described yet. Here, we have found that, ALT-C increased cell migration in MDA-MB-231 at lower concentration (10 nM) and it decreased cell migration at higher concentrations (40, 100 and 1000 nM). ALT-C was able to inhibit MMP-9 activity in human breast cancer (MDA-MB-231) conditioned medium and MMP-2 activity in fibroblastas and human microvascular endothelial cells (HMEC-1) conditioned medium. ALT-C also modulated the expression of angiogenic genes such as VEGF, c-MYC, MMP-2 and MMP-9 and it was able to inhibit transendothelial migration of MDA-MB-231 cells at all concentrations (10, 40, 100 and 1000 nM). In conclusion, ALT-C affects the extracellular matrix remodeling by modulating the activity of MMPs and expression of angiogenic genes essential for tumor growth as well as decreased cell migration. / As metaloproteinases de matriz (MMPs) são fatores chave na progressão tumoral, pois participam do remodelamento da matriz extracelular (ECM), liberam citocinas, fatores de crescimento e são reguladas por moléculas da superfície celular (integrinas). As integrinas são os principais receptores de adesão da superfície celular. Elas interagem com proteínas presentes na matriz extracelular conectando as células ao meio no qual estão inseridas possibilitando sua locomoção e a participação em cascatas de sinalização que influenciam a adesão, sobrevivência e a proliferação celular. Importantes inibidores dessas moléculas foram encontrados em venenos de serpentes denominados de desintegrinas. Alternagina-C (ALT-C), uma desintegrina de veneno da serpente Rhinocerophis alternatus, tem afinidade para a integrina α2β1, modula a adesão, migração e a proliferação celular mas não há nenhum estudo publicado sobre sua influência na atividade das MMPs. Nesse estudo, a ALT-C foi capaz de aumentar a migração celular em MDA-MB-231 em baixa concentração (10 nM) e diminuir a migração em concentrações mais elevadas (40, 100 e 1000 nM). ALT-C também inibiu a atividade de MMP-9 em meio condicionado de células de carcinoma de mama (MDAMB- 231) e atividade de MMP-2 em meio condicionado de fibroblastos e células endoteliais microvasculares humanas (HMEC-1). A desintegrina também foi capaz de modular a expressão de genes angiogênicos como VEGF, c-MYC, MMP-2 e MMP-9 e inibir a transmigração das células tumorais através das células endoteliais. Conclui-se que a ALT-C atua no remodelamento da matriz extracelular do microambiente tumoral por modular a atividade de MMPs e a expressão de genes angiogênicos essenciais no crescimento tumoral, bem como diminuindo a migração celular.
263

Ensaio pré-clínico da desintegrina recombinante DisBa-01 na angio-gênese inflamatória induzida por implantes sintéticos em camundongos

Vieira, Puebla Cassini 11 March 2014 (has links)
Angiogenesis and inflammation act simultaneously in several pathophysiological processes. The interactions with the extracellular matrix (ECM) are required in inflammatory angiogenesis. The non-enzymatic disintegrins comprise a family of peptides, low molecular weight, derived from the venom of snakes, which strongly inhibit the functions of integrins. In this study we evaluated the therapeutic effect of disintegrin DisBa-01, derived from the venom of the snake Rhinocerophis alternatus in inflammatory angiogenesis induced by synthetic implants in mice. Treatment with DisBa-01 inhibited the main key components of fibrovascular tissue induced by implants as inflammation and angiogenesis, without changing the fibrogenic component. This experimental model is the implementation of a synthetic matrix in the subcutaneous tissue of the animal, which induces the formation of fibrovascular tissue rich in inflammatory cells, blood vessels and MEC. The model of synthetic implants, also allows the simultaneous evaluation of inflammation, angiogenesis and tissue repair. The inhibitory effects of DisBa-01 were observed by MPO activity (representing activated neutrophils), NAG activity (representing activated macrophage), the chemokines CXCL -1, MCP-1 and the cytokine TNF- α. The anti-angiogenic activity was observed through the reduction of hemoglobin content, number of vessels, blood flow intra-implant by laser doppler and the levels of VEGF and FGF cytokines. The evaluation of DisBa-01 in multiple parameters provides additional evidence to the therapeutic potential of this disintegrin. / A angiogênese e a inflamação atuam simultaneamente em diversos processos fisiopatológicos. A angiogênese inflamatória é dependente de uma série de interações com a matriz extracelular (MEC) que são mediadas principalmente por receptores integrinas. Assim, peptídeos que possam atuar sobre as integrinas vêm sendo amplamente estudados. Alguns peptídeos como as desintegrinas funcionam como antagonistas das integrinas. As desintegrinas compreendem uma família de polipeptídeo não enzimáticos, de baixo peso molecular, derivados a partir da peçonha de serpentes, que inibem fortemente as funções das integrinas. Neste estudo avaliou-se o efeito terapêutico da desintegrina DisBa-01, derivada da peçonha da serpente Rhinocerophis alternatus na angiogênese inflamatória induzida por implantes sintéticos em camundongos. Este modelo experimental consiste na implantação de uma matriz sintética no tecido subcutâneo do animal, que induz a formação de tecido fibrovascular rico em células inflamatórias, vasos sanguíneos e MEC. O modelo de implantes sintéticos, também permite a avaliação concomitante da inflamação, angiogênese e do reparo tecidual. O tratamento intra-implante com diferentes doses de DisBa-01 inibiu os principais componentes inflamatórios e angiogênicos do tecido fibrovascular. Os efeitos anti-inflamatórios da DisBa-01 foram observados por reduções na atividade de MPO (representando neutrófilos ativados), NAG (representando macrófagos ativados), nas quimiocinas CXCL-1, MCP-1 e pela citocina TNF-α. A atividade anti-angiogênica foi observada através da redução do conteúdo de hemoglobina, do número de vasos em cortes histológicos, bem como por uma redução no fluxo sanguíneo intra-implante através da imagem de perfusão por laser doppler e dos níveis das citocinas pró-angiogênicas VEGF e FGF. No entanto, o tratamento não foi capaz de produzir efeitos significativos sobre os principais componentes do reparo tecidual, como o colágeno e a citocina TGF-β. A avaliação de DisBa-01 nos múltiplos parâmetros reveladas neste estudo demonstrou evidências adicionais ao potencial terapêutico desta desintegrina. / Mestre em Biologia Celular e Estrutural Aplicadas
264

Avaliação da Milk Fat Globule Epidermal Growth Factor 8 (MFG-E8), da integrina αvβ3 e da Leukemia Inhibitory Factor (LIF) na implantação embrionária humana : estudo em modelo in vitro e no endométrio de mulheres com e sem endometriose

Schmitz, Carla Regina January 2015 (has links)
Base teórica: O processo de implantação do embrião no ser humano é extremamente complexo e, ao mesmo tempo, essencial para que a mulher possa engravidar. Neste processo, em que o endométrio precisa sofrer uma série de mudanças para tornar-se receptivo, a adequada expressão de MFG-E8 (milk fat globule epidermal growth factor 8), seu receptor a integrina αvβ3 e LIF (leukemia inhibitory factor) parecem ter um papel importante. Além do mais, mulheres com infertilidade e endometriose podem apresentar a falha de implantação como uma grande barreira para obter seu sucesso terapêutico. Objetivos: Avaliar o papel de MFG-E8 e do seu receptor integrina αvβ3 em um modelo de implantação in vitro com uma linhagem celular trofoblástica e outra de epitélio endometrial. Comparar a expressão de MFG-E8, de integrina αvβ3 e de LIF no endométrio de pacientes férteis e inférteis com endometriose durante a janela de implantação. Métodos: No primeiro ensaio, utilizando-se uma linhagem celular bem diferenciada de adenocarcinoma de endométrio (células Ishikawa) e uma linhagem de coriocarcinoma de trofoblasto, o modelo in vitro de implantação humana foi estabelecido. Para investigação do impacto do bloqueio de MFG-E8 e integrina αvβ3, ambas linhagens celulares foram pré-tratadas com anticorpos contra estas proteínas em diferente concentrações antes do ensaio de adesão. No ensaio subsequente, para comparar a expressão de MFG-E8, de integrina αvβ3 e de LIF no endométrio humano, foram realizadas biópsias no período da janela de implantação (LH+7 a LH+10) com cateter de Pipelle. As amostras foram submetidas a imunohistoquímica, e analisadas através do HSCORE. Resultados: Na avaliação in vitro observamos que as células Ishikawa pré-tratadas com anticorpo anti-MFG-E8 causaram diminuição da adesão das esferas Jar dose-dependente. Por outro lado, o pré-tratamento das esferas Jar não resultou em diminuição significativa da adesão. Pré-tratamento com anticorpos anti-integrina αvβ3, tanto de células Ishikawa como de esferas Jar, causaram inibição significativa, dose-dependente, da adesão das esferas. A análise imunohistoquímica das biópsias realizadas durante a janela de implantação mostrou uma expressão aumentada de MFG-E8 em pacientes com endometriose e infertilidade. Além do mais, houve expressão diminuída de LIF no grupo em estudo. Contudo, não houve diferença estatisticamente significativa na expressão de integrina αvβ3 entre os grupos em estudo. Conclusão: Este estudo demonstrou que, quando se bloqueia MFG-E8 ou seu receptor integrina αvβ3 em células Ishikawa em um modelo in vitro, ocorre uma diminuição de adesão das células Jar. Além do mais, bloqueando-se a integrina αvβ3 nas esferas Jar, também ocorre uma diminuição da adesão destas nas células Ishikawa. No entanto, quando estudamos o endométrio in vivo de pacientes com endometriose e infertilidade, encontramos a expressão aumentada de MFG-E8 e diminuída de LIF durante a janela de implantação no endométrio. / Background: The human implantation process is very complex and, at the same time, it is essential for women to achieve pregnancy. In this process, where the human endometrium must go through a lot of changes in order to become receptive, an adequate expression of MFG-E8 (milk fat globule epidermal growth factor 8), integrin αvβ3 and LIF (leukemia inhibitory factor) appear to play an important role. Furthermore, women with endometriosis and infertility may have in their implantation process the key to achieve pregnancy. Objectives: To investigate the role of MFG-E8 and its receptor integrin αvβ3 in the attachment of trophoblast cells to the endometrial epithelium, in an in vitro model. To compare endometrial expression of MFG-E8, integrin αvβ3 and LIF between fertile patients and patients with endometriosis and infertility during the window of implantation. Methods: In our first assay, by using a well-differentiated endometrial adenocarcinoma cell line (Ishikawa cells) and choriocarcinoma human trophoblast cells (Jar cells), an in vitro model mimicking human implantation was established. To investigate the impact of blocking MFG-E8 and integrin αvβ3, the cell lines were pretreated with antibodies against those proteins at different concentrations before the attachment assay. Moreover, to compare endometrial expression of MFG-E8, integrin αvβ3 and LIF, endometrial biopsies were performed during the window of implantation (LH+7 to LH+10) with the Pipelle catheter. The samples were submitted immunochemistry, and analyzed with HSCORE. Results: Pretreatment of Ishikawa cells with anti-MFG-E8 antibody caused a dosedependent and significant inhibition of attachment is our in vitro assay. On the other hand, pretreatment of Jar spheroids did not result in a significant effect on the attachment rate. Pretreatment of Ishikawa cells as well as Jar spheroids with anti-integrin avb3 antibodies resulted in a dose-dependent, significant inhibition of attachment. The immunochemistry analysis of the endometrial biopsies performed during the window of implantation showed increased MFG-E8 expression in patients with endometriosis and infertility. Moreover, there was lower LIF expression in the study group. Conclusion: This study showed that blocking MFG-E8 and its receptor integrin αvβ3 in Ishikawa cells diminishes Jar spheroid attachment in an in vitro model. Moreover, blocking integrin αvβ3 in the trophoblastic cells also diminished their attachment to the Ishikawa monolayer. Nevertheless, when we studied the endometrium of patients with endometriosis and infertility, we saw an increased expression of MFG-E8 and decreased expression of LIF during the window of implantation.
265

Specific roles of epithelial integrins in chemical and physical sensing of the extracellular matrix to regulate cell shape and polarity

Myllymäki, S.-M. (Satu-Marja) 21 September 2015 (has links)
Abstract Integrins are a large family of αβ-heterodimeric cell adhesion receptors of which the cell type specific expression defines the extracellular matrix (ECM) binding properties of different adherent cell types. In addition to various growth factors and their receptors, epithelial morphogenesis is also executed by dynamic changes in the chemical composition and physical properties of the ECM that controls the shape and behavior of the associated cells via integrin mediated adhesion and signalling. Epithelial cell polarity and contractility are central mechanisms of epithelial shape determination and are established upon spatially, mechanically and chemically sensitive integrin signals of the microenvironment. The functional hierarchy between different integrin heterodimers and their ECM ligands in organizing these tasks has not been systematically addressed. In order to study the relative roles of different integrins, we set up a loss-of-function screen of co-expressed integrin subunits in the Madin-Darby canine kidney (MDCK) epithelial cell line. By analyzing MDCK cystogenesis in three-dimensional (3D) ECMs, we were able to establish a model of how epithelial polarity is organized: cell adhesion either by α2β1- or α6β4-integrins defines the orientation of cell polarity and coordinated functions of α2β1- and α3β1-integrins mediate the establishment of epithelial lumens via cavitation and hollowing, respectively. By analyzing the spreading of MDCK cells, we established that epithelial cell contractility is based on synergistic functions of β1-integrins that mediate cell adhesion and αV-integrins that facilitate ECM rigidity sensing. We also discovered that the hemidesmosomal integrin α6 and integrin β4 did not require heterodimerization to be transported to the plasma membrane (PM) and that integrin β4 may support laminin assembly to the basement membrane (BM) independently of integrin α6. / Tiivistelmä Integriinit ovat suuri molekyyliperhe αβ-heterodimeerisiä adheesioreseptoreja. Integriinit ilmentyvät eri tavoin eri solutyypeissä, ja tämä säätelee sitä, miten solut tarttuvat ja reagoivat erilaisiin soluväliaineisiin. Tällä tavalla integriinit ja soluväliaine osallistuvat myös epiteelimorfogeneesiin lukuisten kasvutekijöiden ja niiden reseptoreiden lisäksi. Epiteelimorfogeneesissä etenkin solujen polarisaatio ja solujen supistuminen ovat tärkeitä tapahtumia, joiden ohjaukseen integriinit ja soluväliaine osallistuvat. Tämän tutkimuksen tarkoituksena oli selvittää eri integriinien ja niiden soluväliaineligandien toiminnallista hierarkiaa epiteelimorfogeneesissä, etenkin solujen polarisaatiossa ja supistumisessa. Integriinien keskinäisten roolien selvittämiseksi hiljensimme ilmentyvät integriinialayksiköt yksitellen munuaisen epiteelisolulinjasta RNA-häirinnän avulla. Mallina epiteelimorfogeneesille käytimme hyväksi munuaisepiteelisolujen kykyä muodostaa rakkularakenteita kolmiulotteisessa soluväliaineessa viljeltyinä. Näitä rakenteita analysoimalla pystyimme muodostamaan mallin siitä, miten polarisoitunut epiteelirakenne organisoituu: α2β1- tai α6β4-integriinien välittämä adheesio tarvitaan solujen polariteetin orientoimiseen ja α2β1- ja α3β1-integriinien yhteistoiminta tarvitaan epiteelisen rakkulan tyhjän sisäosan muodostumiseen, joko apoptoosin tai polarisoituneen kalvokuljetuksen kautta. Tutkimalla solujen levittäytymistä jäykälle kaksiulotteiselle alustalle pystyimme määrittämään, että epiteelisolun supistuminen pohjautuu β1-integriinien välittämän adheesion ja αV-integriinien välittämän väliaineen jäykkyyttä aistivien signaalien yhteistoimintaan. Havaitsimme myös, että hemidesmosomaalisten integriinien α6 ja β4 sekretioon ei tarvittu näiden keskinäistä heterodimerisaatiota ja integriini β4:llä saattaa olla integriini α6:sta riippumaton rooli laminiinin kokoamisessa tyvikalvoon.
266

An integrin required for the encapsulation immune response in the tobacco hornworm, Manduca sexta L. (Lepidoptera: Sphingidae).

Levin, David Michael January 1900 (has links)
Doctor of Philosophy / Department of Entomology / Michael R. Kanost / James R. Nechols / Cellular encapsulation is the immune response in which insects protect themselves from multicellular parasites such as nematodes or parasitoids. During an encapsulation episode, certain insect hemocytes become attracted to a foreign invader and aggregate on its surface. In short order, the invading entity will become entrapped within a capsule comprised of thousands of hemocytes, thus rendering the parasite harmless to the insect host. Although the process of cellular encapsulation has been known for a great many years, very little knowledge yet exists regarding the biochemistry underlying capsule formation. It would seem likely that cell surface adhesion proteins mediate this immune response. In a series of in vivo encapsulation assays in the tobacco hornworm, Manduca sexta, a collection of anti-hemocyte monoclonal antibodies (mAbs) was screened for their ability to inhibit cellular encapsulation. Two of the mAbs that inhibited this immune response and incidentally specifically bind plasmatocytes, MS13 and MS34, were used to isolate a ≈ 90 kDa protein. Several short peptide sequences contained within this protein were acquired via Edman degradation. Degenerate primers based on two of these peptide sequences and total RNA from M. sexta hemocytes were used to perform RT-PCR and 5´ and 3´ RACE. This resulted in a full-length cDNA sequence of 2426 bp. A 2301 bp open reading frame within this cDNA sequence codes for a protein of 767 residues. This protein, denominated [Beta]Ms1, exhibits significant sequence homology to the [Beta]-subunits of integrins, which are a family of transmembrane, heterodimeric glycoproteins that possess adhesive properties. Analysis of recombinant segments of [Beta]Ms1 showed that the protein produced from the PCR product is the antigen to MS13 and MS34 and that these mAbs bind to the region of the integrin that contains the extracellular binding site. Northern blot analysis of various M. sexta tissues together with immunofluorescence labeling with MS13 and MS34 shows that [Beta]Ms1 is solely expressed in plasmatocytes. The totality of these experiments demonstrates that integrins are essential for the cellular immune response of encapsulation.
267

Étude des mécanismes d'action anti-tumorale du collagène XIX / Study of collagen XIX anti-tumor mechanisms

Oudart, Jean-Baptiste 21 October 2015 (has links)
Le collagène XIX est un collagène mineur retrouvé dans la zone de certaines membranes basales spécialisées. Notre laboratoire a montré que son domaine NC1(XIX) est une matrikine présentant des propriétés anti-tumorales et anti-angiogéniques. L'objectif de ce travail est d'étudier les mécanismes aboutissant à cette activité anti-tumorale.Lors de l'invasion tumorale, les cellules cancéreuses dégradent la membrane basale permettant la production de matrikines anti-tumorales. Dans un premier temps, nous avons démontré que la plasmine, qui est une des enzymes clé de l'invasion tumorale, clivait l'extrémité C-terminale du collagène XIX, in vitro et ex vivo, et libérait un peptide présentant une séquence proche du peptide NC1(XIX). Une analyse en modélisation moléculaire a retrouvé que ce peptide adoptait localement la même conformation en coude β de type I que le peptide NC1(XIX). Nous avons ensuite montré que ce peptide inhibait la migration des cellules tumorales in vitro et la croissance tumorale in vivo. Ce travail démontre que le collagène XIX est un nouveau substrat de la plasmine. Ce mécanisme pourrait constituer un moyen de défense de l'organisme contre l'invasion tumorale.Les principaux récepteurs des matrikines appartiennent à la famille des intégrines. Dans un modèle de mélanome humain (cellules SK-MEL-28), nous avons caractérisé l'intégrine αvβ3, comme récepteur du peptide NC1(XIX). Nous avons également démontré que la fixation du peptide NC1(XIX) sur l'intégrine αvβ3 induisait une diminution des phosphorylations de FAK sur le résidu de tyrosine 861, de la sous unité p85 de la PI3K sur le résidu de tyrosine 458, de PDK1 sur le résidu de sérine 241, d'Akt sur les résidus de thréonine 308 et de sérine 473, de mTOR sur les résidus de sérine 2448 et 2481, et de GSK3β sur le résidu de sérine 9. L'inhibition de cette voie FAK / PI3K / Akt / mTOR, largement impliquée dans la transduction du signal dans le mélanome peut, en partie, expliquer les effets anti-tumoraux du peptide NC1(XIX).Parallèlement, ce travail nous a permis de mettre au point différentes techniques de PCR, de Western blot et d'ELISA pour quantifier l'expression du collagène XIX et de son peptide NC1(XIX). Cette étape constitue un préalable essentiel pour une application éventuelle en biologie clinique. / Type XIX collagen is a minor collagen localized in specialized basement membranes. Our laboratory demonstrated that its NC1(XIX) domain is a matrikine which presents anti-tumor and anti-angiogenic properties. The aim of this work was to study the mechanisms leading to this anti-tumor activity.During tumor invasion, cancer cells degrade basement membrane components leading to anti-tumor matrikine release. First, we demonstrated that plasmin, a key enzyme in tumor invasion, cleaved the C-terminal domain of type XIX collagen, in vitro and ex vivo, and released a peptide with a sequence in close vicinity to the NC1(XIX) peptide. Molecular modeling studies showed that NC1(XIX) peptide and the released fragment adopted locally the same type I β-turn conformation. Then, we showed that this peptide inhibited migration of tumor cells in vitro and tumor growth in vivo. This study demonstrated that collagen XIX is a novel proteolytic substrate for plasmin. Such release may constitute a defense of the organism against tumor invasion.The main matrikine receptors belong to the integrin family. In a model of human melanoma cells (SK-MEL-28), we characterized αvβ3 integrin as NC1(XIX) peptide receptor. We also demonstrated that the binding of NC1(XIX) peptide on αvβ3 integrin induced a decrease in phosphorylation of FAK tyrosine 861 residue, PI3K p85 tyrosine 458, PDK1 serine 241,Akt threonine 308 and serine 473, mTOR serine 2448 and 2481, and GSK3β serine 9. The decreased activity of this FAK / PI3K / Akt / mTOR pathway, heavily involved in melanoma signal transduction, could explain, at least in part, the antitumor effects of NC1(XIX) peptide.Meanwhile, this work allowed us to develop PCR, Western blot and ELISA to quantify the expression of collagen XIX and its NC1(XIX) peptide. These steps were required for clinical applications.
268

Hematopoietic stem cells in co-culture with mesenchymal stromal cells - modeling the niche compartments in vitro

Ordemann, Rainer, Jing, Duohui, Fonseca, Ana-Violeta, Alakel, Nael, Fierro, Fernando A., Muller, Katrin, Bornhauser, Martin, Ehninger, Gerhard, Corbeil, Denis 04 January 2016 (has links)
Background Hematopoietic stem cells located in the bone marrow interact with a specific microenvironment referred to as the stem cell niche. Data derived from ex vivo co-culture systems using mesenchymal stromal cells as a feeder cell layer suggest that cell-to-cell contact has a significant impact on the expansion, migratory potential and ‘stemness’ of hematopoietic stem cells. Here we investigated in detail the spatial relationship between hematopoietic stem cells and mesenchymal stromal cells during ex vivo expansion. Design and Methods In the co-culture system, we defined three distinct localizations of hematopoietic stem cells relative to the mesenchymal stromal cell layer: (i) those in supernatant (non-adherent cells); (ii) those adhering to the surface of mesenchymal stromal cells (phase-bright cells) and (iii) those beneath the mesenchymal stromal cells (phase-dim cells). Cell cycle, proliferation, cell division and immunophenotype of these three cell fractions were evaluated from day 1 to 7. Results Phase-bright cells contained the highest proportion of cycling progenitors during co-culture. In contrast, phase-dim cells divided much more slowly and retained a more immature phenotype compared to the other cell fractions. The phase-dim compartment was soon enriched for CD34+/CD38− cells. Migration beneath the mesenchymal stromal cell layer could be hampered by inhibiting integrin β1 or CXCR4. Conclusions Our data suggest that the mesenchymal stromal cell surface is the predominant site of proliferation of hematopoietic stem cells, whereas the compartment beneath the mesenchymal stromal cell layer seems to mimic the stem cell niche for more immature cells. The SDF-1/CXCR4 interaction and integrin-mediated cell adhesion play important roles in the distribution of hematopoietic stem cells in the co-culture system.
269

Control by CCM complex of the dialog between integrins and cadherins for the vascular stability / Régulation par le complexe CCM du dialogue entre intégrines et cadhérines pour le maintien de la stabilité vasculaire.

Lisowska, Justyna 24 November 2014 (has links)
Les interactions cellule-cellule et cellule-matrice extracellulaire (MEC) sont cruciales pour entretenir la cohésion tissulaire. Ces deux types d'adhésions sont fonctionnellement interconnectés par un dialogue permanent qui met en jeu des voies de signalisation convergentes régulant notamment l'architecture et la contractilité du cytosquelette d'acto-myosine sous-jacent. Ce dialogue permet d'établir un équilibre de forces intracellulaires en réponse à la tension appliquée par le milieu extérieur. L'endothélium des vaisseaux sanguins est un tissu soumis à des conditions mécaniques particulières. En plus des compressions intercellulaires subies par tout épithélium, les cellules endothéliales (CEs) doivent également subir et résister aux forces hémodynamiques du flux sanguin et à la rigidité de la lame basale – deux signaux mécaniques agissant de part et d'autre de l'endothélium. Les Cerebral Cavernous Maformations (CCM) ou encore angiomes caverneux sont des lésions vasculaires hémorragiques d'origine génétique qui se développent au niveau des capillaires du système nerveux central et qui se caractérisent par des défauts dans l'environnement proche des CEs. La perte des jonctions intercellulaires et du recouvrement par les cellules murales, l'organisation aberrante de la membrane basale aussi que la stagnation du flux sanguin sont les caractéristiques des CCM. C'est pourquoi nous avons choisi cette pathologie comme modèle intéressant de mécanotransduction mettant en jeu le dialogue entre les intégrines et les cadhérines. En effet, les trois gènes indifféremment mutés dans cette pathologie codent pour des protéines, CCM1-3, qui s'associent en un complexe ternaire et qui sont reconnues comme des acteurs importants de la régulation des jonctions adhérentes. Des études moléculaires et protéomiques montrant que le complexe CCM interagit avec la protéine ICAP-1, un régulateur négatif de l'intégrine β1, nous ont conduit à formuler l'hypothèse selon laquelle ce complexe jouerait un rôle pivot dans la signalisation croisée entre ces intégrines et cadhérines. Les études effectuées pendant ma thèse ont démontré que les protéines CCM régulent l'homéostasie tensionnelle médiée par les structures d'adhérence intercellulaires et à la MEC par leur action inhibitrice sur l'intégrine β1 et en controlant une balance d'activité entre les deux isoformes de ROCK, ROCK1 et ROCK2. Nous avons montré que, suite à la perte des protéines CCMs, la suractivation de l'intégrine β1 augmente la sensibilité des CEs aux signaux mécaniques comme la rigidité de la MEC ou les forces hémodynamiques du flux sanguin. Il en résulte une suractivation de la contractilité cellulaire dépendante de ROCK1 déclenchant une boucle de rétrocontrôle mécanique conduisant à l'amplification des tensions intra- et extracellulaire et brisant ainsi l'homéostasie tensionnelle pour favoriser le phénotype malin. / Cell-cell or cell-matrix interactions have crucial roles in the maintenance of the physical cohesion of any tissue. In addition, growing body of evidence indicates that these two adhesion systems do not act independently, but rather are functionally interconnected by a permanent crosstalk. This dialog usually operates via common molecules that trigger convergent signaling as well as by actomyosin network which, by providing physical link, contributes to establishment of intracellular force counterbalancing tension applied by extracellular surrounding. Blood vessels endothelium is a particular tissue in term of mechanical conditions. Apart from intracellular compression, endothelial lining needs to resist hemodynamic forces as well as rigidity of the basal membrane - two mechanical inputs acting from opposite sides of the endothelial layer. Cerebral Cavernous Malformation (CCM) is a sporadically acquired or inherited disease of venous capillaries within neuro-vascular unit characterized by defects in all aspects of local microenvironment. Loss of intra-endothelial junctions and mural cell coverage, aberrant organization of basal lamina as well as stagnant blood flow are features of CCM lesions. Thereby, CCM became for us an interesting model to study mechanotrasduction process and in this context, the cross-talk between integrin and cadherin mediated adhesion structures. Indeed, CCM proteins are well recognized players involved in a control of VE-cadherin mediated intracellular junctions. In addition, CCM1 was found to interact with ICAP-1, a negative regulator of β1 integrin, raising the possibility that this complex most likely acts as molecular node regulating β1 integrin/ VE-cadherin convergent signaling pathways.Studies performed during this thesis have demonstrated that CCM complex coordinates cadherin- and integrin-mediated tensional homeostasis by repressing β1 integrin activation and maintaining a balance of activity between the two isoforms of RhoA-associated kinases ROCK1 and ROCK2. We have found that β1 integrin sustained over-activation upon CCM proteins loss contributes to increased ECs sensitivity to mechanical cues, such as ECM physical reorganization or hemodynamic force that in turn activates ROCK1-dependent contractility. This establishes a positive feedback mechanical loop that breaks tensional homeostasis and switches on the malignant phenotype.
270

Role of Tissue-Resident Memory T (TRM) Cells in CD8+ T Cell Immunity and Response to Anti-PD-1 Immunotherapy : Involvement of TGF-β and αV Integrins / Rôle des cellules T mémoires résidentes dans le tissu (TRM) dans l’immunité T CD8 et la réponse aux immunothérapies ciblant PD-1 : implication du TGF-β et des intégrines αV dans leur formation

Malenica, Ines 25 July 2019 (has links)
La survie des patients atteints de cancer et traités avec des thérapies conventionnelles reste faible dans plusieurs types de tumeurs. Récemment, une nouvelle approche immunothérapeutique a été développée pour cibler le système immunitaire au lieu de la tumeur elle-même, afin de restaurer la fonctionnalité des cellules immunitaires et la destruction des cellules cancéreuses. L’immunothérapie ciblant le récepteur inhibiteur PD-1 occupe une place privilégiée dans les thérapies anticancéreuses en raison de sa haute spécificité et de sa faible toxicité par rapport aux thérapies conventionnelles. Cependant, le taux de réponse reste faible avec seulement 20 à 25% de patients répondant à une immunothérapie anti-PD-1. Il est donc important de comprendre les mécanismes associés à la résistance à ces thérapies et d’identifier des biomarqueurs prédictifs de réponse. L'expression du ligand de PD-1, PD-L1, sur les cellules tumorales, la charge mutationnelle tumorale et l'infiltration tumorale par les lymphocytes ont déjà été décrits, mais de nouveaux biomarqueurs sont nécessaires pour mieux déterminer la sous-population de patients susceptible de bénéficier de ces traitements. Au cours de ce travail, nous avons établi une cohorte de 118 patients atteints d'un cancer du poumon non à petites cellules (CBNPC) traités avec une immunothérapie anti-PD-1/PD-L1, et nous avons étudié l'expression de plusieurs biomarqueurs potentiels, en particulier les cellules T mémoires résidentes dans le tissu (TRM) CD8+CD103+. Ces cellules constituent un candidat potentiel car elles représentent une population privilégiée de lymphocytes T CD8 grâce à l’expression de PD-1 et une forte capacité cytotoxique vis-à-vis des cellules tumorales autologues suite à la neutralisation de l’interaction de PD-1 avec PD-L1. Nous montrons qu’une forte infiltration de tumeurs de CBNPC avec des cellules TRM corrèle à une survie sans progression plus élevée (PFS) et une réponse plus efficace à anti-PD-1 que les tumeurs avec une faible infiltration par des TRM. De plus, les tumeurs qui expriment fortement ICAM-1, un ligand de l’intégrine LFA-1 exprimée sur les lymphocytes T CD8, sont hautement infiltrées par des TRM. Par ailleurs, il est bien connu que le signal TGF-β est crucial pour l’induction de CD103 et la formation de TRM CD8+CD103+. Je me suis donc intéressée à l'activation du TGF-β par les intégrines αV exprimée par les cellules tumorales humaines et murines. À l'aide des modèles in vitro et in vivo, nous montrons que les cellules tumorales exprimant les intégrines αV activent le TGF-β et induisent l'expression de CD103 à la fois par les cellules T CD8+ provenant de cellules mononucléées du sang périphérique (PBMC) et de lymphocytes infiltrant la tumeur (TIL). L’expression plus faible de CD103 par les TIL CD8+ de souris greffées avec des tumeurs déficientes pour l’expression d'αV n'a pas d'effet sur le contrôle de la croissance tumorale. De manière intéressante, nous montrons dans des modèles de tumeurs déficientes pour l’expression d’αV, que le traitement avec des anticorps anti-PD-1 bloquants corrèle avec un meilleur contrôle de la croissance tumorale et une meilleure réponse à l'immunothérapie anti-PD-1 qui sont associés à une infiltration plus forte de TIL et un état d'activation plus élevé des TIL CD8+ exerçant une activité cytotoxique spécifique. De plus, une expression élevée de l'intégrine αV dans les tumeurs corrèle avec une réponse plus faible des patients atteints de CBNPC à une immunothérapie anti-PD-1/PD-L1. Ces données montrent comment trois marqueurs distincts, cellules TRM, ICAM-1 et les intégrines αV, régulent le microenvironnement tumoral et l’immunité T CD8 avec des implications potentielles pour potentialiser les réponses aux immunothérapies. / The survival of cancer patients treated with conventional therapies remains low in multiple cancers. Recently, a new immunotherapeutic approach has been developed to target the immune system instead of the tumor itself, in order to restore immune cell functions in cancer destruction. Immunotherapy targeting the T cell inhibitory receptor PD-1 occupies a privileged place in cancer therapy thanks to its high specificity and low toxicity compared to chemotherapies. However, the response rate remains low with only 20-25% of patients responding to anti-PD-1 immunotherapy. An important issue is therefore to understand the mechanisms associated with resistance to these therapies and to identify the predictive biomarkers of response. The expression of the PD-1 ligand, PD-L1, on tumor cells, tumor mutational burden (TMB) and tumor infiltration by lymphocytes have been described to predict the response to immune checkpoint blockade (ICB). However, new biomarkers are needed to better determine patient subpopulation which could benefit from this treatment. To address this question, we established a cohort of 118 non-small cell lung cancer (NSCLC) patients treated with anti-PD-1/PD-L1 immunotherapy and studied the expression of several potential biomarkers. Tissue-resident memory T (TRM) cells are a potential candidate because they represent a distinct population of CD8+ T cells highly expressing integrin αEβ7 (CD103) and PD-1; and showing strong cytotoxic capacity towards autologous tumor cells upon neutralisation of PD-1/PD-L1 interaction. Results from the present study show that high infiltration of TRM cells in NSCLC tumors correlates with higher progression-free survival (PFS) and a better response to anti-PD-1/PD-L1 immunotherapy. Moreover, tumors with high expression levels of ICAM-1, the ligand of integrin LFA-1 expressed on T cells, show higher TRM infiltration. TGF-β is a cytokine directly involved in CD103 induction on activated tumor-specific T cells. Therefore, I also investigated the role of αV integrins in activating TGF-β and thereby in controlling TRM differentiation and anti-tumor T cell immunity. Using human and mouse models, we show that tumor cells expressing αV integrins activate TGF-β, which can in turn induce expression of CD103 on CD8+ T cells in vitro on peripheral blood mononuclear cells (PBMCs) and in vivo on tumor infiltrating lymphocytes (TIL). However, lower CD103 expression on CD8+ TIL and thus CD103+ TRM cell formation in C57BL/6 mice engrafted with αV-lacking cancer cells had no effect on tumor growth control. Remarkably, αV-deficient tumors responded more effectively to anti-PD-1 immunotherapy than αV-efficient tumors and this response correlates with higher tumor infiltration by activated CD8+ T cells and stronger cytotoxic activity toward autologous cancer cells. Moreover, high expression of αV integrins in NSCLC tumors correlates with worse response to anti-PD-1/PD-L1 immunotherapy. These data show how three distinct markers, TRM cells, ICAM-1, and αV integrins regulate the tumor microenvironment and CD8+ T cell immunity, with potential implications in improving response to ICB immunotherapies.

Page generated in 0.0404 seconds