• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 65
  • 19
  • 15
  • 9
  • 3
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 145
  • 44
  • 34
  • 32
  • 22
  • 21
  • 21
  • 19
  • 19
  • 19
  • 16
  • 16
  • 15
  • 15
  • 14
  • 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.
41

Μελέτη του ρόλου του αυξητικού παράγοντα HARP στην παθοφυσιολογία του ανθρώπινου προστάτη. / Study on the role of growth factor HARP in the pathophysiology of the human prostate.

Χατζηαποστόλου, Μαρία 24 June 2007 (has links)
Η εγκαθίδρυση και ανάπτυξη καρκίνου του προστάτη διαμεσολαβείται από τη δράση μιας πλειάδας ογκογενετικών αυξητικών παραγόντων. Μέχρι σήμερα έχει αναδειχθεί η εμπλοκή του αυξητικού παράγοντα HARP στην ανάπτυξη καρκινικών όγκων διαφορετικής προελεύσεως. Στην παρούσα εργασία, διερευνήθηκε η πιθανή συμμετοχή της HARP στην ανάπτυξη καρκίνου του προστάτη. Με εφαρμογή μιας αντινοηματικής στρατηγικής πραγματοποιήθηκε καταστολή έκφρασης της HARP στην καρκινική κυτταρική σειρά προστάτη LNCaP και μελετήθηκε τόσο ο ρόλος της HARP στην αύξηση και μεταναστευτική ικανότητα των καρκινικών κυττάρων, όσο και η ενδεχόμενη αγγειογενετική δράση της in vitro και in vivo. Η εξωγενώς χορηγούμενη ανασυνδυασμένη HARP ανθρώπου, ήταν μιτογόνος για τα κύτταρα LNCaP. Επιπρόσθετα η καταστολή έκφρασης της ενδογενούς HARP, ανέδειξε την αναγκαιότητα του συγκεκριμένου αυξητικού παράγοντα για τη μετανάστευση των κυττάρων LNCaP καθώς και για την κυτταρική αύξηση τόσο σε συνθήκες εξαρτώμενες όσο και ανεξάρτητες από την προσκόλληση σε υπόστρωμα. Οι επαγόμενες, από τα κύτταρα LNCaP, λειτουργίες των ενδοθηλιακών κυττάρων in vitro και ο σχηματισμός νέων αγγείων in vivo, αναχαιτίστηκαν όταν ανεστάλη η έκφραση της HARP. Ο αυξητικός παράγοντας ινοβλαστών FGF-2 είναι ένας πλειοτροπικός αυξητικός παράγοντας, ο οποίος διαδραματίζει σημαντικό ρόλο στην εγκαθίδρυση και ανάπτυξη καρκίνου του προστάτη. Τα αποτελέσματα της παρούσας διατριβής, κατέδειξαν ότι ο FGF-2 δύναται να επάγει σε σημαντικό ποσοστό τον πολλαπλασιασμό και τη μετανάστευση των κυττάρων LNCaP. Το μόριο της HARP φαίνεται να μεσολαβεί προκειμένου να εκδηλωθούν οι διεγερτικές δράσεις του FGF-2, δεδομένου ότι τελευταίος δεν επηρέασε αντίστοιχες λειτουργίες των κυττάρων LNCaP στα οποία είχε κατασταλεί η έκφραση της HARP. Επιπλέον, ο FGF-2 διέγειρε την έκφραση και έκκριση της HARP από τα κύτταρα LNCaP και αύξησε τη δραστηριότητα λουσιφεράσης πλασμιδιακού οχήματος αναφοράς, στο οποίο είχε κλωνοποιηθεί η ρυθμιστική περιοχή του γονιδίου της HARP. Ο ειδικός αναστολέας του υποδοχέα FGFR-1, SU-5402, αναχαίτισε την επαγόμενη από τον FGF-2 ενεργοποίηση του γονιδίου της HARP και την επακόλουθη έκκριση της πρωτεΐνης, οδηγώντας με τον τρόπο αυτό σε εξασθένιση του κυτταρικού πολλαπλασιασμού. Επώαση των κυττάρων LNCaP με πυροσταφυλικό νάτριο, το οποίο απομακρύνει με έμμεσο τρόπο το υπεροξείδιο του υδρογόνου, ανέδειξε την εξάρτηση των διεγερτικών δράσεων του FGF-2 από την ενδοκυτταρική παραγωγή υπεροξειδίου του υδρογόνου, ενώ ανάσχεση της δραστικότητας του FGFR-1 ανέστειλε τον επαγόμενο από τον FGF-2 σχηματισμό δραστικών μορφών οξυγόνου. Με χρησιμοποίηση ολιγονουκλεοτιδικών δολωμάτων έναντι του ΑΡ-1 και εφαρμογή κατευθυνόμενης μεταλλαξιγένεσης στη ρυθμιστική περιοχή του γονιδίου της HARP, διαπιστώθηκε η εμπλοκή του ΑΡ-1 στην επαγόμενη από τον FGF-2 έκφραση και έκκριση της HARP. Η επίδραση του FGF-2 στα κύτταρα LNCaP, φαίνεται να οφείλεται στη δέσμευση των Fra-1, JunD και της ενεργού μορφής της c-Jun στη ρυθμιστική περιοχή του γονιδίου της HARP. Συμπερασματικά, καταδεικνύεται ο σημαντικός ρυθμιστικός ρόλος του αυξητικού παράγοντα HARP σε ποικίλες βιολογικές διεργασίες των καρκινικών κυττάρων ανθρώπινου προστάτη. Επιπλέον, στην παρούσα εργασία προτείνεται ο ρόλος και ο μηχανισμός δράσης του αυξητικού παράγοντα FGF-2 στα κύτταρα LNCaP, ενώ ταυτόχρονα αντικατοπτρίζεται η πολυπλοκότητα των μονοπατιών αυξητικών παραγόντων που εμπλέκονται στον καρκίνο του προστάτη. / The development and growth of human prostate cancer is mediated by many tumor cell-derived growth factors. Heparin affin regulatory peptide (HARP) seems to be involved in the progression of several tumors of diverse origin. In the present work, we sought to determine if HARP is implicated in human prostate cancer. An antisense strategy for inhibition of HARP expression in the human prostate cancer cell line LNCaP was used to study the role of HARP on cancer cell growth, migration and angiogenic potential in vitro and in vivo. Exogenous human recombinant HARP was mitogenic for LNCaP cells. By decreasing the expression of endogenous HARP, we found that HARP was essential for LNCaP cell migration, as well as anchorage-dependent and independent growth. Endothelial cell functions in vitro and blood vessel formation in vivo induced by LNCaP cells were also inhibited when HARP expression was diminished. Fibroblast growth factor 2 (FGF-2) is a pleiotropic growth factor that has been implicated in prostate carcinoma formation and progression. In the present study we found that exogenous FGF-2 significantly increased human prostate cancer LNCaP cell proliferation and migration. HARP seems to be an important mediator of FGF-2 stimulatory effects, since the latter had no effect on stably transfected LNCaP cells that did not express HARP. Moreover, FGF-2 significantly induced HARP expression and secretion by LNCaP cells and increased luciferase activity of a reporter gene vector carrying the full length promoter of HARP gene. The FGFR1-specific inhibitor SU-5402 blocked the FGF-2-increased HARP gene activation and the consequent protein release, leading to impairment of LNCaP cell proliferation. Treatment of LNCaP cells with the hydrogen peroxide scavenger pyruvate, pointed to the dependence of FGF-2-induced HARP expression and LNCaP cell proliferation on hydrogen peroxide generation, and blockade of FGFR1 activity abrogated the FGF-2-induced production of reactive oxygen species. Activator protein-1 (AP-1) seems to be involved in FGF-2-stimulated HARP expression and secretion by LNCaP cells, as revealed using AP-1 decoy oligonucleotides and point mutation analyses in the HARP gene promoter. Binding of AP-1 complexes consisting of Fra-1, JunD and phospho-c-Jun, to the HARP promoter seems to be amenable for FGF-2 effect. These results point to an important regulator role of HARP in diverse biological activities in human prostate cancer cells. Furthermore, the present work establishes the role and the mode of activity of FGF-2 in LNCaP cells and reflects the many-sidedness of growth factor pathways within prostate cancer.
42

Fibroblast growth factor-2 protects neonatal rat cardiac myocytes from doxorubicin-induced damage via protein kinase C- dependent effects on efflux drug transporters

Wang, Jie 22 January 2013 (has links)
Introduction: Therapeutic agents like doxorubicin, an anthracycline antibiotic drug, are widely used in cancer chemotherapy. The use of doxorubicin is limited however by an increased risk of cardiac damage as a side effect, and an increased cancer cell drug resistance mediated by efflux drug transporters. Strategies are needed to protect the heart and still allow the benefits of drug treatment. “Basic” fibroblast growth factor-2 (FGF-2) is a multi-functional protein. It is angiogenic and cardioprotective against ischemia-reperfusion injury. FGF-2 can also regulate cancer cell drug resistance or sensitivity, however, so far, there is no evidence that FGF-2 protects against doxorubicin-induced cardiac damage through effects on efflux drug transporter levels or function. Aims: To investigate whether: (1) FGF-2 can increase resistance to doxorubicin-induced neonatal rat cardiac myocyte damage; and if so whether (2) an effect on efflux drug transporters might contribute to this cardioprotection by FGF-2. Methods: Neonatal rat cardiac myocyte cultures were treated with doxorubicin in the absence or presence of pre-treatment with FGF-2. To assess cell damage: (i) culture medium was tested for lactate dehydrogenase (LDH) activity as an indication of plasma membrane disruption; (ii) cells were stained with fluorescent apoptosis and necrosis biomarkers as well as (iii) terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and acridine orange to assess DNA fragmentation or compaction. The role of FGF receptor (FGFR) or protein kinase C (PKC) was addressed through use of inhibitors including SU5402, or chelerythrine as well as bisindomaleimide. Multidrug resistance gene 1a and 1b (MDR1a, 1b), multidrug resistance gene 2 (MDR2) and multidrug resistance-related protein 1 (MRP1) gene expression, as well as the function of MDRs and MRPs protein products were assessed by real-time reverse transcriptase-polymerase chain reaction (qPCR), as well as retention/extrusion of (fluorescent) doxorubicin/calcein in cardiac myocytes, respectively. Efflux drug transporter inhibitors, including 20 µM cyclosporine A (CsA), 2 µM verapamil and 1 µM Tariquidar (XR9576) were used to asssess for a direct effect of FGF-2 on transporter function. Fluorescence-activated cell sorting (FACS) was used to measure fluorescent doxorubicin/calcein levels inside treated cardiac myocytes. Results: Doxorubicin increased the incidence of programmed cell death, DNA damage, and lysosome and LDH activity, while decreasing cell number at 24 hours. FGF-2 prevented the detrimental effects of doxorubicin. In turn, the protective effects of FGF-2 were blocked in the presence of FGFR or PKC inhibitors. FGF-2 treatment significantly increased MDR1a, MDR1b, MDR2, MRP1 RNA levels by qPCR, and protein levels as assessed by function, and specifically extrusion of doxorubicin/calcein, in the presence of doxorubicin when compared to doxorubicin treatment alone. Furthermore, inhibition of efflux drug transporters with CsA and Tariquidar (XR9576) significantly reduced the ability of FGF-2 to protect against doxorubicin-induced damage; the beneficial effect of FGF-2 was completely blocked by pretreatment with verapamil. Conclusion(s): These data indicate for the first time that exogenous FGF-2 can increase resistance to doxorubicin-induced neonatal rat cardiac myocyte damage, and implicate PKC and regulation of efflux transporter protein levels and/or function in the mechanism.
43

Fibroblast growth factor-2 protects neonatal rat cardiac myocytes from doxorubicin-induced damage via protein kinase C- dependent effects on efflux drug transporters

Wang, Jie 22 January 2013 (has links)
Introduction: Therapeutic agents like doxorubicin, an anthracycline antibiotic drug, are widely used in cancer chemotherapy. The use of doxorubicin is limited however by an increased risk of cardiac damage as a side effect, and an increased cancer cell drug resistance mediated by efflux drug transporters. Strategies are needed to protect the heart and still allow the benefits of drug treatment. “Basic” fibroblast growth factor-2 (FGF-2) is a multi-functional protein. It is angiogenic and cardioprotective against ischemia-reperfusion injury. FGF-2 can also regulate cancer cell drug resistance or sensitivity, however, so far, there is no evidence that FGF-2 protects against doxorubicin-induced cardiac damage through effects on efflux drug transporter levels or function. Aims: To investigate whether: (1) FGF-2 can increase resistance to doxorubicin-induced neonatal rat cardiac myocyte damage; and if so whether (2) an effect on efflux drug transporters might contribute to this cardioprotection by FGF-2. Methods: Neonatal rat cardiac myocyte cultures were treated with doxorubicin in the absence or presence of pre-treatment with FGF-2. To assess cell damage: (i) culture medium was tested for lactate dehydrogenase (LDH) activity as an indication of plasma membrane disruption; (ii) cells were stained with fluorescent apoptosis and necrosis biomarkers as well as (iii) terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and acridine orange to assess DNA fragmentation or compaction. The role of FGF receptor (FGFR) or protein kinase C (PKC) was addressed through use of inhibitors including SU5402, or chelerythrine as well as bisindomaleimide. Multidrug resistance gene 1a and 1b (MDR1a, 1b), multidrug resistance gene 2 (MDR2) and multidrug resistance-related protein 1 (MRP1) gene expression, as well as the function of MDRs and MRPs protein products were assessed by real-time reverse transcriptase-polymerase chain reaction (qPCR), as well as retention/extrusion of (fluorescent) doxorubicin/calcein in cardiac myocytes, respectively. Efflux drug transporter inhibitors, including 20 µM cyclosporine A (CsA), 2 µM verapamil and 1 µM Tariquidar (XR9576) were used to asssess for a direct effect of FGF-2 on transporter function. Fluorescence-activated cell sorting (FACS) was used to measure fluorescent doxorubicin/calcein levels inside treated cardiac myocytes. Results: Doxorubicin increased the incidence of programmed cell death, DNA damage, and lysosome and LDH activity, while decreasing cell number at 24 hours. FGF-2 prevented the detrimental effects of doxorubicin. In turn, the protective effects of FGF-2 were blocked in the presence of FGFR or PKC inhibitors. FGF-2 treatment significantly increased MDR1a, MDR1b, MDR2, MRP1 RNA levels by qPCR, and protein levels as assessed by function, and specifically extrusion of doxorubicin/calcein, in the presence of doxorubicin when compared to doxorubicin treatment alone. Furthermore, inhibition of efflux drug transporters with CsA and Tariquidar (XR9576) significantly reduced the ability of FGF-2 to protect against doxorubicin-induced damage; the beneficial effect of FGF-2 was completely blocked by pretreatment with verapamil. Conclusion(s): These data indicate for the first time that exogenous FGF-2 can increase resistance to doxorubicin-induced neonatal rat cardiac myocyte damage, and implicate PKC and regulation of efflux transporter protein levels and/or function in the mechanism.
44

The Role of Fgf and Its Downstream Effectors in Otic and Epibranchial Development in Zebrafish

Padanad, Mahesh 2011 August 1900 (has links)
In vertebrates, the otic placode forms inner ear and epibranchial placodes produce sensory ganglia within branchial clefts. Fibroblast growth factor (FGF) family of protein ligands from the surrounding tissues are responsible for otic and epibranchial placode induction. Members of pax2/5/8 family of transcription factors function as mediators during otic induction. To understand the temporal and spatial requirements of Fgf and their interaction with pax2/8 for otic induction, we used heat shock inducible transgenic lines of zebrafish to misexpress fgf3/8 and pax2a/8 under the control of hsp70 promoter. Loss of function studies were done to examine the functions of pax2/8 genes in regulating otic and epibranchial development. We show that global transient activation of hs:fgf3 or hs:fgf8 at mid-late gastrula stages (7-8 hpf) severely impairs otic induction, in part by disrupting formation of the principal signaling centers in the hindbrain. Additionally, mosaic studies show that high-level misexpression blocks otic fate cell-autonomously, whereas low to moderate levels promote otic development. At later stages high-level Fgf misexpression, both globally and locally does not inhibit otic fate, but rather causes a dramatic expansion of endogenous otic domains. Misexpression of hs:pax2a or hs:pax8 also expands endogenous otic domains but is not sufficient to bypass the requirement for Fgf signaling. Co-misexpression of Fgf with pax2a or pax8 leads to production of ectopic otic tissue in a broad range of cranial ectoderm. These data show that changes in timing, distribution and level of Fgf signaling and its downstream effectors influences otic induction. We show that otic and epibranchial placodes are induced at different times and by distinct mechanisms. Initially, Fgf from surrounding tissues induces otic expression of pax8 and sox3, which cooperate synergistically to establish otic fate. Subsequently, pax8 along with pax2a/pax2b downregulate foxi1 expression in otic cells, which is necessary for further otic development. Additionally, pax2/8 activate otic expression of fgf24, which induces epibranchial expression of sox3. Blocking functions of fgf24 or sox3 causes severe epibranchial deficiencies but has little effect on otic development. These results support the model whereby the otic placode forms first and induces epibranchial placodes through pax2/8-dependent Fgf24 signaling.
45

Inhibition of IGF-I and B-FGF Stimulated Growth of Human Retinal Endothelial Cells by the Somatostatin Analogue, Octreotide: A Potential Treatment for Ocular Neovascularization

Grant, Maria B., Caballero, Sergio, Millard, William J. 20 October 1993 (has links)
No description available.
46

Strategies to improve the biological performance of protein therapeutics / Strategien zur Verbesserung der biologischen Wirkung von Proteintherapeutika

Gador, Eva January 2018 (has links) (PDF)
During the last decades the number of biologics increased dramatically and several biopharmaceutical drugs such as peptides, therapeutic proteins, hormones, enzymes, vaccines, monoclonal antibodies and antibody-drug conjugates conquered the market. Moreover, administration and local delivery of growth factors has gained substantial importance in the field of tissue engineering. Despite progress that has been made over the last decades formulation and delivery of therapeutic proteins is still a challenge. Thus, we worked on formulation and delivery strategies of therapeutic proteins to improve their biological performance. Phase I of this work deals with protein stability with the main focus on a liquid protein formulation of the dimeric fusion protein PR-15, a lesion specific platelet adhesion inhibitor. In order to develop an adequate formulation ensuring the stability and bioactivity of PR-15 during storage at 4 °C, a pH screening, a forced degradation and a Design of Experiments (DoE) was performed. First the stability and bioactivity of PR-15 in 50 mM histidine buffer in relation to pH was evaluated in a short-term storage stability study at 25 °C and 40 °C for 4 and 8 weeks using different analytical methods. Additionally, potential degradation pathways of PR-15 were investigated under stressed conditions such as heat treatment, acidic or basic pH, freeze-thaw cycles, light exposure, induced oxidation and induced deamidation during the forced degradation study. Moreover, we were able to identify the main degradation product of PR-15 by performing LC/ESI-MS analysis. Further optimization of the injectable PR 15 formulation concerning pH, the choice of buffer and the addition of excipients was studied in the following DoE and finally an optimal PR-15 formulation was found. The growth factors BMP-2, IGF-I and TGF-β3 were selected for the differentiation of stem cells for tissue engineering of cartilage and bone in order to prepare multifunctionalized osteochondral implants for the regeneration of cartilage defects. Silk fibroin (SF) was chosen as biomaterial because of its biocompatibility, mechanical properties and its opportunity for biofunctionalization. Ideal geometry of SF scaffolds with optimal porosity was found in order to generate both tissues on one scaffold. The growth factors BMP-2 and IGF-I were modified to allow spatially restricted covalent immobilization on the generated porous SF scaffolds. In order to perform site-directed covalent coupling by the usage of click chemistry on two opposite sides of the scaffold, we genetically engineered BMP-2 (not shown in this work; performed by Barbara Tabisz) and IGF-I for the introduction of alkyne or azide bearing artificial amino acids. TGF β3 was immobilized to beads through common EDC/NHS chemistry requiring no modification and distributed in the pores of the entire scaffold. For this reason protein modification, protein engineering, protein immobilization and bioconjugation are investigated in phase II. Beside the synthesis the focus was on the characterization of such modified proteins and its conjugates. The field of protein engineering offers a wide range of possibilities to modify existing proteins or to design new proteins with prolonged serum half-life, increased conformational stability or improved release rates according to their clinical use. Site-directed click chemistry and non-site-directed EDC/NHS chemistry were used for bioconjugation and protein immobilization with the aim to underline the preferences of site-directed coupling. We chose three strategies for the incorporation of alkyne or azide functionality for the performance of click reaction into the protein of interest: diazonium coupling reaction, PEGylation and genetic engineering. Azido groups were successfully introduced into SF by implementation of diazonium coupling and alkyne, amino or acid functionality was incorporated into FGF-2 as model protein by means of thiol PEGylation. The proper folding of FGF-2 after PEGylation was assessed by fluorescence spectroscopy, WST-1 proliferation assay ensured moderate bioactivity and the purity of PEGylated FGF-2 samples was monitored with RP-HPLC. Moreover, the modification of native FGF-2 with 10 kDa PEG chains resulted in enhanced thermal stability. Additionally, we genetically engineered one IGF-I mutant by incorporating the unnatural amino acid propargyl-L-lysine (plk) at position 65 into the IGF-I amino acid sequence and were able to express hardly verifiable amounts of plk-IGF-I. Consequently, plk-IGF-I expression has to be further optimized in future studies in order to generate plk-IGF-I with higher yields. Bioconjugation of PEGylated FGF-2 with functionalized silk was performed in solution and was successful for click as well as EDC/NHS chemistry. However, substantial amounts of unreacted PEG-FGF-2 were adsorbed to SF and could not be removed from the reaction mixture making it impossible to expose the advantages of click chemistry in relation to EDC/NHS chemistry. The immobilization of PEG-FGF-2 to microspheres was a trial to increase product yield and to remove unreacted PEG-FGF-2 from reaction mixture. Bound PEG-FGF-2 was visualized by fluorescence imaging or flow cytometry and bioactivity was assessed by analysis of the proliferation of NIH 3T3 cells. However, immobilization on beads raised the same issue as in solution: adsorption caused by electrostatic interactions of positively charged FGF-2 and negatively charged SF or beads. Finally, we were not able to prove superiority of site-directed click chemistry over non-site-directed EDC/NHS. The skills and knowledge in protein immobilization as well as protein characterization acquired during phase II helped us in phase III to engineer cartilage tissue in biofunctionalized SF scaffolds. The approach of covalent immobilization of the required growth factors is relevant because of their short in vivo half-lives and aimed at controlling their bioavailability. So TGF-β3 was covalently coupled by means of EDC/NHS chemistry to biocompatible and biostable PMMA beads. Herein, we directly compared bioactivity of covalently coupled and adsorbed TGF-β3. During the so-called luciferase assay bioactivity of covalent coupled as well as adsorbed TGF-β3 on PMMA beads was ensured. In order to investigate the real influence of EDC/NHS chemistry on TGF-β3’s bioactivity, the amount of immobilized TGF-β3 on PMMA beads was determined. Therefore, an ELISA method was established. The assessment of total amount of TGF-β3 immobilized on the PMMA beads allowed as to calculate coupling efficiency. A significantly higher coupling efficiency was determined for the coupling of TGF-β3 via EDC/NHS chemistry compared to the reaction without coupling reagents indicating a small amount of adsorbed TGF-β3. These results provide opportunity to determine the consequence of coupling by means of EDC/NHS chemistry for TGF β3 bioactivity. At first sight, no statistically significant difference between covalent immobilized and adsorbed TGF-β3 was observed regarding relative luciferase activities. But during comparison of total and active amount of TGF-β3 on PMMA beads detected by ELISA or luciferase assay, respectively, a decrease of TGF-β3’s bioactivity became apparent. Nevertheless, immobilized TGF β3 was further investigated in combination with SF scaffolds in order to drive BMSCs to the chondrogenic lineage. According to the results obtained through histological and immunohistochemical studies, biochemical assays as well as qRT-PCR of gene expression from BMSCs after 21 days in culture immobilized TGF-β3 was able to engineer cartilage tissue. These findings support the thesis that local presentation of TGF β3 is superior towards exogenous TGF β3 for the development of hyaline cartilage. Furthermore, we conclude that covalent immobilized TGF β3 is not only superior towards exogenously supplemented TGF-β3 but also superior towards adsorbed TGF-β3 for articular hyaline cartilage tissue engineering. Diffusion processes were inhibited through covalent immobilization of TGF-β3 to PMMA beads and thereby a stable and consistent TGF-β3 concentration was maintained in the target area. With the knowledge acquired during phase II and III as well as during the studies of Barbara Tabisz concerning the expression and purification of plk-BMP-2 we made considerable progress towards the formation of multifunctionalized osteochondral implants for the regeneration of cartilage defects. However, further studies are required for the translation of these insights into the development of multifunctionalized osteochondral SF scaffolds. / In den letzten Jahrzehnten stieg die Zahl der Biologika dramatisch an und mehrere biopharmazeutische Arzneimittel wie Peptide, therapeutische Proteine, Hormone, Enzyme, Impfstoffe, monoklonale Antikörper und Antikörper-Wirkstoff-Konjugate eroberten den Markt. Darüber hinaus hat die Applikation und lokale Verabreichung von Wachstumsfaktoren im Bereich des Tissue Engineerings eine wesentliche Bedeutung erlangt. Trotz der in den letzten Jahrzehnten erzielten Fortschritte ist die Formulierung und Verabreichung therapeutischer Proteine noch immer eine Herausforderung. Daher haben wir uns in dieser Arbeit mit der Formulierung und Verabreichung therapeutischer Proteine beschäftigt und Strategien entwickelt, um deren biologische Wirkung zu verbessern. In Phase I dieser Arbeit konzentrieren wir uns auf die Stabilität des dimeren Fusionsproteins PR 15, einem Inhibitor der Adhäsion von Plättchen an arterielle Gefäßläsionen. Um eine geeignete flüssige Formulierung zu entwickeln, welche die Stabilität und Bioaktivität von PR-15 während der Lagerung bei 4 °C sicherstellt, wurde ein pH Screening, eine Forced Degradation Studie und ein Design of Experiments (DoE) durchgeführt. Zuerst wurde die Stabilität und Bioaktivität von PR-15 bei verschiedenen pH Werten in 50 mM Histidinpuffer in einer Kurzzeitstabilitätsstudie bei 25 °C und 40 °C nach 4 und 8 Wochen mit Hilfe verschiedener analytischer Methoden beobachtet. Des Weiteren wurden mögliche Abbauwege von PR-15 unter Stressbedingungen wie erhöhter Temperatur, saurem oder basischem pH-Wert, Einfrier-Auftau-Zyklen, Lichteinwirkung, induzierter Oxidation sowie induzierter Deamidierung während der Forced Degradation Studie untersucht. Darüber hinaus konnten wir das Hauptabbauprodukt von PR-15 durch LC/ESI-MS Analysen identifizieren. Im folgenden DoE wurde die injizierbare PR-15 Formulierung weiter optimiert und bezüglich pH, der Wahl des Puffers sowie der Zugabe von Hilfsstoffen analysiert, bis letztendlich eine optimale PR 15-Formulierung gefunden wurde. Die Wachstumsfaktoren BMP-2, IGF-I und TGF-β3 wurden zur Differenzierung von Stammzellen für das Tissue Engineering von Knochen und Knorpel ausgewählt, um multifunktionalisierte osteochondrale Implantate zur Regeneration von Knorpeldefekten herzustellen. Seidenfibroin (SF) wurde aufgrund seiner Biokompatibilität, seiner mechanischen Eigenschaften und seiner Möglichkeiten zur Biofunktionalisierung als Biomaterial gewählt. Zuerst wurden SF-Scaffolds mit idealer Geometrie und optimaler Porosität erzeugt, um sowohl Knochen also auch Knorpel auf einem Scaffold herzustellen. Um eine räumlich begrenzte kovalente Immobilisierung der Wachstumsfaktoren BMP-2 und IGF-I auf den porösen SF-Scaffolds zu ermöglichen, wurden diese mit unnatürlichen Aminosäuren genetisch modifiziert. Das Einführen von Alkin- bzw. Azidresten in die Aminosäuresequenz von BMP-2 (in dieser Arbeit nicht gezeigt; von Barbara Tabisz durchgeführt) und IGF-I erlaubt unter Verwendung der Click-Chemie eine ortsgerichtete kovalente Kopplung der Wachstumsfaktoren an zwei gegenüberliegenden Seiten der Scaffolds. TGF-β3 wurde durch gewöhnliche EDC/NHS-Chemie, welche keine Modifikation erforderte, kovalent an Mikrosphären immobilisiert und in den Poren des gesamten SF-Scaffolds verteilt. Daher beschäftigen wir uns in Phase II mit der Modifikation von Proteinen, dem Protein Engineering, der Immobilisation von Proteinen und mit Biokonjugation. Neben der Synthese lag der Fokus auf der Charakterisierung modifizierter Proteine und deren Konjugaten. Das Gebiet des Protein Engineerings bietet eine Vielzahl von Möglichkeiten, bestehende Proteine zu modifizieren oder neue Proteine mit verlängerter Serumhalbwertszeit, erhöhter konformativer Stabilität oder verbesserten Freisetzungsraten entsprechend der klinischen Anwendung zu entwickeln. Die ortsspezifische Click-Chemie und die nicht-ortsspezifische EDC/NHS-Chemie wurden für die Biokonjugation und die Immobilisierung von Proteinen verwendet mit dem Ziel, die Vorzüge der ortsgerichteten Kopplung hervorzuheben. Für den Einbau der für die Durchführung der Click-Reaktion erforderlichen Alkin- bzw. Azidfunktionalität in das betreffende Protein wurden drei Strategien ausgewählt: die Azokupplung, die PEGylierung und die gentechnische Modifizierung. Azidgruppen wurden mittels Azokupplung erfolgreich in SF eingebaut und die Alkin-, Amino- oder Säurefunktionalität wurde mittels PEGylierung der Cysteine in das Modellprotein FGF-2 integriert. Die korrekte Faltung von FGF-2 nach erfolgreicher PEGylierung wurde durch Fluoreszenzspektroskopie bestätigt, im WST-1 Proliferationsassay wurde eine angemessene Bioaktivität festgestellt und die Reinheit von PEGylierten FGF-2 wurde mittels RP-HPLC analysiert. Darüber hinaus führte die Modifikation von nativem FGF-2 mit 10 kDa PEG-Ketten zu einer erhöhten thermischen Stabilität. Des Weiteren wurde ein IGF-I-Mutant gentechnisch hergestellt, indem die unnatürliche Aminosäure Propargyl-L-Lysin (Plk) an Position 65 in die IGF-I-Sequenz eingebaut wurde. Da letztendlich lediglich kaum nachweisbare Mengen an Plk-IGF-I exprimiert werden konnten, muss die Plk-IGF-I-Expression in anschließenden Studien weiter optimiert werden, um Plk-IGF-I mit höheren Ausbeuten erzeugen zu können. Die Biokonjugation von PEGyliertem FGF-2 und funktionalisierter Seide wurde sowohl mittels Click- als auch mittels EDC/NHS-Chemie erfolgreich durchgeführt. Allerdings wurden erhebliche Mengen PEG-FGF-2 lediglich an SF adsorbiert und nicht kovalent gekoppelt und konnten schlussendlich nicht aus dem Reaktionsgemisch entfernt werden. Die anschließende Immobilisierung von PEG-FGF-2 an Mikrosphären, war ein Versuch die Ausbeute der Reaktion zu erhöhen und adsorbiertes PEG-FGF-2 leichter zu entfernen. Immobilisiertes PEG-FGF-2 wurde mittels Fluoreszenzmikroskopie und/oder Durchflusszytometrie nachgewiesen und die Bioaktivität wurde durch die Analyse der Proliferation von NIH-3T3-Zellen ermittelt. Die Immobilisierung auf Mikrosphären führte jedoch zu demselben Problem wie in Lösung: Adsorption von positiv geladenem FGF-2 an negativ geladenes SF bzw. negativ geladenen Mikrosphären durch elektrostatische Wechselwirkungen. Schließlich waren wir nicht in der Lage, die Überlegenheit der ortsgerichteten Click-Chemie gegenüber der nicht-ortsgerichteten EDC/ NHS-Chemie zu beweisen. Die während Phase II erworbenen Fähigkeiten und Kenntnisse im Bereich der Immobilisierung und Charakterisierung von Proteinen halfen uns in Phase III Knorpelgewebe in biofunktionalisierten SF-Scaffolds zu erzeugen. Der Ansatz der kovalenten Immobilisierung, der für das Tissue Engineering von Knorpel erforderlichen Wachstumsfaktoren, ist aufgrund ihrer kurzen in vivo Halbwertszeiten von Bedeutung und zielt darauf ab, ihre Bioverfügbarkeit zu kontrollieren. So wurde TGF-β3 mittels EDC/NHS-Chemie kovalent an biokompatible und biostabile PMMA-Mikrosphären gekoppelt. Mit Hilfe des sogenannten Luciferase-Assays wurden die Bioaktivitäten von kovalent gekoppeltem sowie von adsorbiertem TGF-β3 auf PMMA-Mikrosphären ermittelt. Um die Kopplungseffizienz zu berechnen und den tatsächlichen Einfluss der EDC/NHS-Chemie auf die Bioaktivität von TGF-β3 zu untersuchen, wurde die Menge an immobilisiertem TGF-β3 auf PMMA-Mikrosphären mittels ELISA bestimmt. Für die Kopplung von TGF-β3 mittels EDC/NHS-Chemie wurde eine signifikant höhere Kopplungseffizienz im Vergleich zu der Reaktion ohne Kopplungsreagenzien, welche eine geringe Menge an adsorbiertem TGF-β3 zeigte, bestimmt. Bei alleiniger Betrachtung der Ergebnisse des Luciferase-Assays, bei welchem kein statistisch signifikanter Unterschied zwischen kovalent immobilisiertem und adsorbiertem TGF-β3 bezüglich der relativen Luciferase-Aktivität beobachtet wurde, scheint es als hätte die EDC/NHS-Kopplung keinen Einfluss auf die Bioaktivität von TGF β3. Beim Vergleich der mittels ELISA bestimmten TGF β3 Gesamtmenge und der mittels Luciferase-Assay bestimmten Menge an aktivem TGF-β3 auf den PMMA-Mikrosphären, wurde jedoch ein Verlust der Bioaktivität von TGF-β3 durch die EDC/NHS-Kopplung deutlich. Ungeachtet dessen, wurde immobilisiertes TGF-β3 genutzt, um Knorpelgewebe in SF-Scaffolds aus BMSCs zu generieren. Nach den Ergebnissen der histologischen und immunhistochemischen Untersuchungen, der biochemischen Assays sowie der qRT-PCR der Genexpression von BMSCs nach 21 Tagen in Kultur, gelang es uns unter Verwendung von immobilisiertem TGF-β3 Knorpelgewebe aufzubauen. Diese Ergebnisse unterstützen die These, dass die lokale Präsentation von TGF-β3 gegenüber exogen zugegebenem TGF-β3 für die Entwicklung von hyalinem Knorpel überlegen ist. Außerdem schließen wir daraus, dass kovalent immobilisiertes TGF-β3 nicht nur gegenüber exogen zugegebenem TGF-β3 für die Entwicklung von hyalinem Knorpelgewebe überlegen ist, sondern auch gegenüber adsorbiertem TGF-β3. Diffusionsprozesse konnten durch kovalente Immobilisierung von TGF-β3 an PMMA-Mikrosphären verhindert werden und damit eine stabile und gleichmäßige TGF β3-Konzentration am Wirkort aufrechterhalten werden. Mit den in Phase II und III gewonnenen Erkenntnissen und den Untersuchungen von Barbara Tabisz zur Expression und Aufreinigung von plk-BMP-2 haben wir erhebliche Fortschritte bei der Entwicklung multifunktionaler osteochondraler Implantate zur Regeneration von Knorpeldefekten gemacht. Für die Umsetzung dieser Erkenntnisse zur Herstellung multifunktionaler osteochondraler SF-Scaffolds sind jedoch weitere Studien erforderlich.
47

THE ROLE OF THE ETS TRANSCRIPTION FACTOR Elf5 IN LUNG DEVELOPMENT

METZGER, DAVID EDWARD January 2007 (has links)
No description available.
48

Régulateurs traductionnels de l'expression génique de la différenciation et du stress cellulaire / Translational regulation during the differentiation and cellular stress

Hantelys, Fransky 20 February 2017 (has links)
La cellule est susceptible de modifier l'expression de ces gènes en fonction de son environnement. Dans les cellules eucaryotes, la régulation de l'expression de ces gènes se présente dans plusieurs étapes. Cette régulation peut intervenir dès la transcription de l'ADN jusqu'au devenir des transcrits. La régulation post-transcriptionnelle tient un rôle déterminant dans la synthèse protéique. Elle regroupe l'ensemble des contrôles qui s'exercent sur les transcrits. Cette régulation est induite en réponse à différents stimuli comme la différenciation ou lors de stress cellulaires. En situation de stress, la traduction canonique dépendante de la coiffe est bloquée, à l'exception de certains ARNm essentiels pour assurer la survie des cellules. De ce fait, les cellules mettent en place un mécanisme alternatif afin de continuer la traduction. Un des mécanismes de traduction, implique le site d'entrée interne du ribosome ou IRES (Internal Ribosome Entry Site). L'IRES est une séquence en structure secondaire dans la partie 5' non-traduite de certains ARNm. Il existe des facteurs responsables de leur activation appelés ITAF ou IRES-transacting factor, permettant le recrutement des ribosomes pour initier la traduction. Les protéines pouvant se lier aux ARN sont les acteurs majeurs de l'activation des IRES. Mon travail de thèse est d'étudier les régulateurs post-transcriptionnels en réponse à différents stimuli par le biais de la traduction IRES-dépendante. Dans la première partie de mon projet, nous avons montré la régulation de la traduction via l'activation de l'IRES du FGF1 et ce de manière promoteur-dépendante au cours de la différenciation des myoblastes. Grâce à la technique de résonance plasmonique de surface (SPR) nous avons découvert deux protéines p54nrb/NONO et hnRNPM en tant qu'ITAF capables de former un complexe pour activer l'IRES du FGF1 durant la différenciation des myoblastes. Dans la deuxième partie de ma thèse, nous avons démontré l'existence de l'IRES du VEGFD durant un choc thermique dans les cellules cancéreuses. Nous avons aussi découvert que cette activation est médiée par un ITAF qui est la nucléoline, jamais démontrée auparavant comme ITAF de l'IRES du VEGFD. D'après nos résultats, le stress thermique induit la délocalisation de la nucléoline du noyau vers le cytoplasme pour changer la conformation de l'IRES du VEGFD afin de continuer sa traduction. Dans la troisième partie de mon projet, j'ai étudié de manière générale la régulation des gènes angiogéniques et lymphangiogéniques. L'ensemble des données montre que ces gènes sont majoritairement régulés au niveau traductionnel dans les cardiomyocytes en condition hypoxique. En étudiant les IRES angiogéniques et lymphangiogéniques, nos résultats montrent l'activation de ces IRES à différents temps au cours de l'hypoxie précoce. Dans la même condition, nous avons découvert la protéine vasohibin-1 comme ITAF hypoxique et spécifique de l'IRES du FGF1 dans les cardiomyocytes. En conclusion, nous avons découvert différents ITAF spécifiques à un IRES et en fonction du stress. P54nrb/NONO, hnRNPM sont des ITAF de l'IRES du FGF1 durant la différenciation cellulaire et la vasohibine-1 en hypoxie dans les cardiomyocytes. La nucléoline permet d'activer un IRES du VEGFD en réponse au choc thermique. / In cell, gene expression can be modified depending on the cellular microenvironment. Regulation of gene expression occurs at different levels, ranging from the transcription of the DNA to the mRNA. Among the post- transcriptional regulation, the control of translation plays a crucial role. In particular, the translational regulation occurs in response to different stimuli such as cell differentiation or cell stress. In stress condition, the canonical cap-dependent translation is blocked, excepted some mRNAs that are translated by alternative mechanisms. One of these mechanisms involves the structural elements of the mRNAs, the IRES (Internal Ribosome Entry Sites). The IRES activation involves some factors called ITAFs (IRES trans-acting factors), which allow the internal recruitement of ribosomes to initiate translation. My thesis is to study the mechanisms of IRES-dependent translation regulation in response to different stimuli, and to identify ITAFs responsible for this regulation. In the first part of my project, we have shown that the translation controlled by the FGF1 mRNA IRES is activated. This activation depends on its own promoter during the early phase of murine myoblast differentiation. Through biomolecular interaction analysis technology by surface plasmon resonance coupled to mass spectrometry (BIA/MS), we identified two proteins, p54nrb/NONO and hnRNPM bound both to the IRES and the FGF1gene promoter. These two proteins are both ITAFs activators of IRES and activators of FGF1 promoter transcription, resulting in a coupling of transcription and translation responsible for the induction of the FGF1 expression during myoblast differentiation. In the second part of this thesis, we demonstrated the existence of an IRES within the VEGFD mRNA. This IRES is activated by heat shock in mammary murine carcinoma. BIA/MS technology has enabled us to identify nucleolin as ITAF responsible for this activation. SHAPE experiments revealed the presence of two alternative structures of VEGFD IRES. According to our results, the heat shock induced the relocation of nucleolin from the nucleus to the cytoplasm, suggesting its binding to the mRNA in the cytoplasm could stabilize the conformation of the mRNA VEGFD IRES and activate its translation. The third part of my thesis focused on translational regulation of lymphangiogenic and angiogenic genes into cardiomyocytes in hypoxic conditions. The data obtained by the semi-global approach Fluidigm indicate that only few genes are induced at the transcriptional level, while the majority of them, especially those which have the mRNA IRES, are activated at translational level in hypoxic cardiomyocytes. I have also shown that the mRNA IRES of factors (lymph)angiogenic VEGF and FGF are activated during early hypoxia. Through Technology BIA/MS, I identified a specific hypoxic ITAF of FGF1 IRES in cardiomyocytes: it is the vasohibin - 1 protein involved in angiogenesis and stress tolerance. So, my thesis has enabled to make progress in understanding the mechanisms of IRES-dependent translation regulation. In addition, I have demonstrated that in cardiomyocytes during hypoxia the gene expression is surprisingly regulated at translational level. My work led to the identification of several molecular actors responsible for the regulation of mRNA (lymph)angiogenic factors translation, which could play a key role in ischemic pathologies and in cancer, and provide new targets therapeutic.
49

Interaction entre les voies de signalisation FGF et Notch lors de la migration de la parapineale dans le cerveau asymétrique du poisson zèbre / Crosstalk between FGF and Notch signaling pathways during the collective migration of parapineal cells in the left right asymmetric zebrafish brain

Wei, Lu 26 November 2018 (has links)
Lors du développement de l'asymétrie gauche droite dans le cerveau du poisson zèbre, un petit groupe de cellules, le parapinéale, migre collectivement depuis la ligne médiane vers la partie gauche de l'épithalamus. Cette migration est défectueuse dans des mutants pour le gène fgf8, indiquant que le facteur Fgf8 (Fibroblast Growth Factor 8), sécrété de part et d'autre de la ligne médiane, est requis pour la migration. Cependant, l'orientation gauche de la migration dépend de l'activation, plus précocement dans l'épithalamus gauche, de la voie de signalisation Nodal/TGFb (Transforming Growth Factor). Par conséquent, la parapinéale est un modèle de choix pour comprendre comment les cellules migrent collectivement en réponse aux Fgf et pour étudier comment d'autres voies de signalisation modulent ce processus. L'imagerie en temps réel d'un transgène rapporteur de la signalisation FGF a révélé que la voie FGF est activée préférentiellement dans quelques cellules de tête, c'est à dire localisées au front de migration. L'expression globale d'un récepteur aux Fgf activé de façon constitutive (CA-FgfR1) interfère avec la migration de la parapinéale en contexte sauvage mais est capable de restaurer à la fois la migration de la parapinéale et l'activation focale de la voie FGF au front de migration dans les mutants fgf8-/-. De plus, l'activation focale de la voie FGF dans seulement quelques cellules de parapinéale est suffisante pour restaurer la migration de tout le collectif dans les mutants fgf8-/-. Finalement, nos données montrent que la signalisation Nodal contribue à restreindre et à biaiser l'activation de la voie FGF afin d'orienter la migration de la parapinéale vers le côté gauche (Manuscript n°1). Par la suite, mes travaux de thèse ont visé à comprendre comment l'activation de la voie FGF est restreinte à quelques cellules, bien que toutes les cellules de parapinéale semblent compétentes pour activer la voie. Nos résultats montrent que la signalisation Notch est capable de restreindre l'activation de la voie FGF. La perte ou le gain de fonction de la voie Notch entrainent respectivement une augmentation ou une diminution de l'activité FGF, associés à des défauts de migration de la parapinéale dans les deux contextes. De plus, la diminution ou l'augmentation artificielle du niveau d'activation de la voie FGF peut respectivement restaurer la migration de la parapinéale ou aggraver les défauts de migration en absence d'activité Notch. Nos données indiquent que la signalisation Notch restreint l'activation de la voie FGF au sein des cellules de parapinéale pour permettre la migration du collectif (Manuscript n°2). La voie Notch est également requise pour la spécification d'un nombre correct de cellules de parapinéale, indépendamment de la voie FGF. En parallèle, nous avons analysé la fonction de MMP2 (Matrix Metalloprotease 2), une protéine exprimée mosaïquement dans la parapinéale et candidate pour moduler la signalisation FGF. Cependant, nous n'avons observé aucun défaut de spécification ou de migration de la parapinéale dans les embryons mutants pour le gène mmp2 -/- (Manuscript n°3). Mon travail de thèse révèle un rôle de la voie Notch pour restreindre l'activation de la signalisation FGF dans quelques cellules de parapinéale, un processus qui est biaisé par la voie Nodal afin d'orienter la migration du collectif vers la gauche. Ces données pourraient permettre de mieux comprendre les interactions entre les voies de signalisation FGF, Notch et Nodal dans d'autres modèles de migration cellulaire collective comme, par exemple, la migration des cellules cancéreuses. / During the establishment of left-right asymmetry in the zebrafish brain, a small group of cells, the parapineal, collectively migrates from the dorsal midline of the epithalamus to the left in most wild-type embryos. Parapineal migration requires Fibroblastic Growth Factor 8 (Fgf8), a secreted signal expressed bilaterally in epithalamic tissues surrounding the parapineal. The left bias in the orientation of parapineal migration depends on the activity of Cyclops, a secreted factor of the Nodal/TGFß family that is transiently expressed in the left epithalamus prior to parapineal migration. Therefore, the parapineal provides a powerful new model to understand FGF dependent collective cell migration and to study how other signaling pathways modulate this process. Live imaging of an FGF reporter transgene revealed that the FGF pathway is activated in only few parapineal cells that are usually located at the leading edge of migration. Global expression of a constitutively activated Fgf receptor (CA-FGFR) delays migration in wild-type, while it partially restores both parapineal migration and focal activation of the FGF reporter transgene in fgf8-/- mutant embryos. Importantly, focal activation of FGF signaling in few parapineal cells is sufficient to restore collective migration in fgf8-/- mutants. Finally, Nodal asymmetry contributes to restrict and left-bias the activation of the FGF pathway (Manuscript n°1). Following this work, my thesis project aimed at understanding how the activation of the FGF pathway is restricted to few cells, despite all parapineal cells apparently being competent to activate the pathway. We showed that Notch signaling is able to restrict FGF activity. Loss or gain of function of the Notch pathway respectively triggers an increase or decrease in FGF activity, which correlate with PP migration defects. Moreover, decreasing or increasing FGF activity levels respectively rescues or aggravates parapineal migration defects in Notch loss-of-function context. Our data indicate that Notch signaling restricts the activation of the FGF pathway within parapineal cells to promote their collective migration (Manuscript n°2). We also found that Notch pathway is required for the specification of a correct number of parapineal cells, independently of FGF pathway. In parallel, we analysed the function of MMP2 (Matrix Metalloprotease 2), a protein mosaïcally expressed in the parapineal and a candidate to modulate FGF signaling. However, we found no significant defects in the specification or migration of parapineal cells in mmp2-/- mutant embryos (Manuscript n°3). My PhD work reveals a role for Notch signaling in restricting the activation of FGF signaling within few parapineal cells, a process that is biased by Nodal pathway to the left and required for the migration of the entire parapineal. These data provide insights into the interaction of FGF, Notch and Nodal/TGFb signaling pathways that may be applicable to other models of collective cell migration, such as cancer cells migration for instance.
50

Größenegulation der Augenanlage von Xenopus laevis durch Inhibition von Hedgehog-, Fgf- und Wnt-Signalen / size-regulation of the Xenopus laevis eye anlage by inhibition of Hedgehog-, Fgf- and Wnt-signals

Cornesse, Yvonne 05 November 2003 (has links)
No description available.

Page generated in 0.0967 seconds