Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Biomater Sci ; 12(12): 3124-3140, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38738995

ABSTRACT

Synthetic tubular grafts currently used in clinical context fail frequently, and the expectations that biomimetic materials could tackle these limitations are high. However, developing tubular materials presenting structural, compositional and functional properties close to those of native tissues remains an unmet challenge. Here we describe a combination of ice templating and topotactic fibrillogenesis of type I collagen, the main component of tissues' extracellular matrix, yielding highly concentrated yet porous tubular collagen materials with controlled hierarchical architecture at multiple length scales, the hallmark of native tissues' organization. By modulating the thermal conductivity of the cylindrical molds, we tune the macroscopic porosity defined by ice. Coupling the aforementioned porosity patterns with two different fibrillogenesis routes results in a new family of tubular materials whose textural features and the supramolecular arrangement of type I collagen are achieved. The resulting materials present hierarchical elastic properties and are successfully colonized by human endothelial cells and alveolar epithelial cells on the luminal side, and by human mesenchymal stem cells on the external side. The proposed straightforward protocol is likely to be adapted for larger graft sizes that address ever-growing clinical needs, such as peripheral arterial disease or tracheal and bronchial reconstructions.


Subject(s)
Biomimetic Materials , Ice , Tissue Engineering , Humans , Biomimetic Materials/chemistry , Porosity , Mesenchymal Stem Cells/cytology , Collagen Type I/chemistry , Animals
2.
Cell Cycle ; 19(14): 1707-1715, 2020 07.
Article in English | MEDLINE | ID: mdl-32552303

ABSTRACT

Budding yeast, Saccharomyces cerevisiae, has been widely used as a model system to study cellular signaling in response to internal and external cues. Yeast was among the first organisms in which the architecture, feedback mechanisms and physiological responses of various MAP kinase signaling cascades were studied in detail. Although these MAP kinase pathways are activated by different signals and elicit diverse cellular responses, such as adaptation to stress and mating, they function as an interconnected signaling network, as they influence each other and, in some cases, even share components. Indeed, various stress signaling pathways interfere with pheromone signaling that triggers a distinct cellular differentiation program. However, the molecular mechanisms responsible for this crosstalk are still poorly understood. Here, we review the general topology of the yeast MAP kinase signaling network and highlight recent and new data revealing how conflicting intrinsic and extrinsic signals are interpreted to orchestrate appropriate cellular responses.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Pheromones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomycetales/metabolism , Signal Transduction , Time Factors
3.
Oxid Med Cell Longev ; 2018: 1240192, 2018.
Article in English | MEDLINE | ID: mdl-29725493

ABSTRACT

BACKGROUND: Severe obstructive sleep apnea (OSA) with chronic intermittent hypoxia (IH) is common in idiopathic pulmonary fibrosis (IPF). Here, we evaluated the impact of IH on bleomycin- (BLM-) induced pulmonary fibrosis in mice. METHODS: C57BL/6J mice received intratracheal BLM or saline and were exposed to IH (40 cycles/hour; FiO2 nadir: 6%; 8 hours/day) or intermittent air (IA). In the four experimental groups, we evaluated (i) survival; (ii) alveolar inflammation, pulmonary edema, lung oxidative stress, and antioxidant enzymes; (iii) lung cell apoptosis; and (iv) pulmonary fibrosis. RESULTS: Survival at day 21 was lower in the BLM-IH group (p < 0.05). Pulmonary fibrosis was more severe at day 21 in BLM-IH mice, as assessed by lung collagen content (p = 0.02) and histology. At day 4, BLM-IH mice developed a more severe neutrophilic alveolitis, (p < 0.001). Lung oxidative stress was observed, and superoxide dismutase and glutathione peroxidase expression was decreased in BLM-IH mice (p < 0.05 versus BLM-IA group). At day 8, pulmonary edema was observed and lung cell apoptosis was increased in the BLM-IH group. CONCLUSION: These results show that exposure to chronic IH increases mortality, lung inflammation, and lung fibrosis in BLM-treated mice. This study raises the question of the worsening impact of severe OSA in IPF patients.


Subject(s)
Bleomycin/adverse effects , Lung Injury/etiology , Sleep Apnea, Obstructive/complications , Animals , Cell Hypoxia , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL
4.
Am J Physiol Lung Cell Mol Physiol ; 314(3): L360-L371, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29167125

ABSTRACT

Distal lung diseases, such as pulmonary fibrosis or acute lung injury, are commonly associated with local alveolar hypoxia that may be deleterious through the stimulation of alveolar epithelial cell (AEC) apoptosis. In various murine models of alveolar injury, administration of allogenic human mesenchymal stem cells (hMSCs) exerts an overall protective paracrine effect, limiting lung inflammation and fibrosis. However, the precise mechanisms on lung cells themselves remain poorly understood. Here, we investigated whether hMSC-conditioned medium (hMSC-CM) would protect AECs from hypoxia-induced apoptosis and explored the mechanisms involved in this cytoprotective effect. Exposure of rat primary AECs to hypoxia (1.5% O2 for 24 h) resulted in hypoxia-inducible factor (HIF)-1α protein stabilization, partly dependent on reactive oxygen species (ROS) accumulation, and in a twofold increase in AEC apoptosis that was prevented by the HIF inhibitor 3-(5'-hydroxymethyl-2'-furyl)-1-benzyl-indazole and the antioxidant drug N-acetyl cysteine. Incubation of AECs with hMSC-CM significantly reduced hypoxia-induced apoptosis. hMSC-CM decreased HIF-1α protein expression, as well as ROS accumulation through an increase in antioxidant enzyme activities. Expression of Bnip3 and CHOP, two proapoptotic targets of HIF-1α and ROS pathways, respectively, was suppressed by hMSC-CM, while Bcl-2 expression was restored. The paracrine protective effect of hMSC was partly dependent on keratinocyte growth factor and hepatocyte growth factor secretion, preventing ROS and HIF-1α accumulation.


Subject(s)
Alveolar Epithelial Cells/cytology , Apoptosis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/physiopathology , Mesenchymal Stem Cells/cytology , Pulmonary Alveoli/cytology , Reactive Oxygen Species/metabolism , Alveolar Epithelial Cells/physiology , Animals , Cells, Cultured , Humans , Male , Mesenchymal Stem Cells/physiology , Pulmonary Alveoli/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction
5.
Am J Physiol Lung Cell Mol Physiol ; 310(5): L439-51, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26702148

ABSTRACT

Administration of bone marrow-derived human mesenchymal stem cells (hMSC) reduces lung inflammation, fibrosis, and mortality in animal models of lung injury, by a mechanism not completely understood. We investigated whether hMSC would prevent epithelial-mesenchymal transition (EMT) induced by hypoxia in primary rat alveolar epithelial cell (AEC). In AEC cultured on semipermeable filters, prolonged hypoxic exposure (1.5% O2 for up to 12 days) induced phenotypic changes consistent with EMT, i.e., a change in cell morphology, a decrease in transepithelial resistance (Rte) and in the expression of epithelial markers [zonula occludens-1 (ZO-1), E-cadherin, AQP-5, TTF-1], together with an increase in mesenchymal markers [vimentin, α-smooth muscle actin (α-SMA)]. Expression of transcription factors driving EMT such as SNAIL1, ZEB1, and TWIST1 increased after 2, 24, and 48 h of hypoxia, respectively. Hypoxia also induced TGF-ß1 mRNA expression and the secretion of active TGF-ß1 in apical medium, and the expression of connective tissue growth factor (CTGF), two inducers of EMT. Coculture of AEC with hMSC partially prevented the decrease in Rte and in ZO-1, E-cadherin, and TTF-1 expression, and the increase in vimentin expression induced by hypoxia. It also abolished the increase in TGF-ß1 expression and in TGF-ß1-induced genes ZEB1, TWIST1, and CTGF. Finally, incubation with human recombinant KGF at a concentration similar to what was measured in hMSC-conditioned media restored the expression of TTF-1 and prevented the increase in TWIST1, TGF-ß1, and CTGF in hypoxic AEC. Our results indicate that hMSC prevent hypoxia-induced alveolar EMT through the paracrine modulation of EMT signaling pathways and suggest that this effect is partly mediated by KGF.


Subject(s)
Alveolar Epithelial Cells/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Mesenchymal Stem Cells/metabolism , Animals , Cell Hypoxia , Cell Line , Connective Tissue Growth Factor/metabolism , Lung/metabolism , Male , Rats, Sprague-Dawley , Signal Transduction/physiology
6.
Am J Physiol Lung Cell Mol Physiol ; 306(11): L975-85, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24682451

ABSTRACT

Mesenchymal stromal cells (MSCs) or their media (MSC-M) were reported to reverse acute lung injury (ALI)-induced decrease of alveolar fluid clearance. To determine the mechanisms by which MSC-M exert their beneficial effects, an in vitro model of alveolar epithelial injury was created by exposing primary rat alveolar epithelial cells (AECs) to hypoxia (3% O2) plus cytomix, a combination of IL-1ß, TNF-α, and IFN-γ. MSC-M were collected from human MSCs exposed for 12 h to either normoxia (MSC-M) or to hypoxia plus cytomix (HCYT-MSC-M). This latter condition was used to model the effect of alveolar inflammation and hypoxia on paracrine secretion of MSCs in the injured lung. Comparison of paracrine soluble factors in MSC media showed that the IL-1 receptor antagonist and prostaglandin E2 were markedly increased while keratinocyte growth factor (KGF) was twofold lower in HCYT-MSC-M compared with MSC-M. In AECs, hypoxia plus cytomix increased protein permeability, reduced amiloride-sensitive short-circuit current (AS-Isc), and also decreased the number of α-epithelial sodium channel (α-ENaC) subunits in the apical membrane. To test the effects of MSC media, MSC-M and HCYT-MSC-M were added for an additional 12 h to AECs exposed to hypoxia plus cytomix. MSC-M and HCYT-MSC-M completely restored epithelial permeability to normal. MSC-M, but not HCYT-MSC-M, significantly prevented the hypoxia plus cytomix-induced decrease of ENaC activity and restored apical α-ENaC channels. Interestingly, KGF-deprived MSC-M were unable to restore amiloride-sensitive sodium transport, indicating a possible role for KGF in the beneficial effect of MSC-M. These results indicate that MSC-M may be a preferable therapeutic option for ALI.


Subject(s)
Alveolar Epithelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Sodium/metabolism , Animals , Apoptosis , Biological Transport , Cell Hypoxia , Cell Membrane Permeability , Cells, Cultured , Culture Media, Conditioned , Dinoprostone/metabolism , Epithelial Sodium Channels/metabolism , Fibroblast Growth Factor 7/metabolism , Humans , Inflammation Mediators/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Male , Paracrine Communication , Pulmonary Alveoli/pathology , Rats , Rats, Sprague-Dawley
7.
Am J Respir Cell Mol Biol ; 50(3): 526-37, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24093724

ABSTRACT

Transepithelial sodium transport via alveolar epithelial Na(+) channels (ENaC) and Na(+),K(+)-ATPase constitutes the driving force for removal of alveolar edema fluid. Alveolar hypoxia associated with pulmonary edema may impair ENaC activity and alveolar Na(+) absorption through a decrease of ENaC subunit expression at the apical membrane of alveolar epithelial cells (AECs). Here, we investigated the mechanism(s) involved in this process in vivo in the ß-Liddle mouse strain mice carrying a truncation of ß-ENaC C-terminus abolishing the interaction between ß-ENaC and the ubiquitin protein-ligase Nedd4-2 that targets the channel for endocytosis and degradation and in vitro in rat AECs. Hypoxia (8% O2 for 24 h) reduced amiloride-sensitive alveolar fluid clearance by 69% in wild-type mice but had no effect in homozygous mutated ß-Liddle littermates. In vitro, acute exposure of AECs to hypoxia (0.5-3% O2 for 1-6 h) rapidly decreased transepithelial Na(+) transport as assessed by equivalent short-circuit current Ieq and the amiloride-sensitive component of Na(+) current across the apical membrane, reflecting ENaC activity. Hypoxia induced a decrease of ENaC subunit expression in the apical membrane of AECs with no change in intracellular expression and induced a 2-fold increase in α-ENaC polyubiquitination. Hypoxic inhibition of amiloride-sensitive Ieq was fully prevented by preincubation with the proteasome inhibitors MG132 and lactacystin or with the antioxidant N-acetyl-cysteine. Our data strongly suggest that Nedd4-2-mediated ubiquitination of ENaC leading to endocytosis and degradation of apical Na(+) channels is a key feature of hypoxia-induced inhibition of transepithelial alveolar Na(+) transport.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Epithelial Cells/enzymology , Epithelial Sodium Channels/metabolism , Hypoxia/enzymology , Proteasome Endopeptidase Complex/metabolism , Pulmonary Alveoli/enzymology , Sodium/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Antioxidants/pharmacology , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Endocytosis , Epithelial Cells/drug effects , Epithelial Sodium Channels/deficiency , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/genetics , Hypoxia/genetics , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucociliary Clearance , Nedd4 Ubiquitin Protein Ligases , Proteasome Inhibitors/pharmacology , Pulmonary Alveoli/drug effects , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Time Factors
8.
PLoS One ; 4(12): e8171, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19997595

ABSTRACT

BACKGROUND: Asymmetric cell divisions are involved in the divergence of the first two lineages of the pre-implantation mouse embryo. They first take place after cell polarization (during compaction) at the 8-cell stage. It is thought that, in contrast to many species, spindle orientation is random, although there is no direct evidence for this. METHODOLOGY/PRINCIPAL FINDINGS: Tubulin-GFP and live imaging with a spinning disk confocal microscope were used to directly study spindle orientation in whole embryos undergoing the 8- to 16-cell stage transition. This approach allowed us to determine that there is no predetermined cleavage pattern in 8-cell compacted mouse embryos and that mitotic spindle orientation in live embryo is only modulated by the extent of cell rounding up during mitosis. CONCLUSIONS: These results clearly demonstrate that spindle orientation is not controlled at the 8- to 16-cell transition, but influenced by cell bulging during mitosis, thus reinforcing the idea that pre-implantation development is highly regulative and not pre-patterned.


Subject(s)
Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Development , Spindle Apparatus/metabolism , Animals , Blastomeres/cytology , Blastomeres/metabolism , Female , Humans , Male , Mice , Mitosis , Time Factors
9.
PLoS One ; 4(9): e7117, 2009 Sep 21.
Article in English | MEDLINE | ID: mdl-19768116

ABSTRACT

BACKGROUND: During mammalian preimplantation development, lineage divergence seems to be controlled by the interplay between asymmetric cell division (once cells are polarized) and positional information. In the mouse embryo, two distinct cell populations are first observed at the 16-cell stage and can be distinguished by both their position (outside or inside) and their phenotype (polarized or non-polarized). Many efforts have been made during the last decade to characterize the molecular mechanisms driving lineage divergence. METHODOLOGY/PRINCIPAL FINDINGS: In order to evaluate the importance of cell polarity in the determination of cell fate we have disturbed the activity of the apical complex aPKC/PAR6 using siRNA to down-regulate aPKClambda expression. Here we show that depletion of aPKClambda results in an absence of tight junctions and in severe polarity defects at the 16-cell stage. Importantly, we found that, in absence of aPKClambda, cell fate depends on the cellular context: depletion of aPKClambda in all cells results in a strong reduction of inner cells at the 16-cell stage, while inhibition of aPKClambda in only half of the embryo biases the progeny of aPKClambda defective blastomeres towards the inner cell mass. Finally, our study points to a role of cell shape in controlling cell position and thus lineage allocation. CONCLUSION: Our data show that aPKClambda is dispensable for the establishment of polarity at the 8-cell stage but is essential for the stabilization of cell polarity at the 16-cell stage and for cell positioning. Moreover, this study reveals that in addition to positional information and asymmetric cell divisions, cell shape plays an important role for the control of lineage divergence during mouse preimplantation development. Cell shape is able to influence both the type of division (symmetric or asymmetric) and the position of the blastomeres within the embryo.


Subject(s)
Blastocyst/cytology , Blastocyst/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Animals , Cell Lineage , Cell Polarity , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Male , Mice , Microscopy, Video/methods , Mitosis , RNA Interference , RNA, Messenger/metabolism , Tight Junctions
10.
Mol Cell Endocrinol ; 282(1-2): 70-7, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-18155829

ABSTRACT

The first 4 days of mouse pre-implantation development are characterized by a period of segmentation, including morphogenetic events that are required for the divergence of embryonic and extra-embryonic lineages. These extra-embryonic tissues are essential for the implantation into the maternal uterus and for the development of the foetus. In this review, we first discuss data showing unambiguously that no essential axis of development is set up before the late blastocyst stage, and explain why the pre-patterning described during the early phases (segmentation) of development in other vertebrates cannot apply to mammalian pre-implantation period. Then, we describe important cellular and molecular events that are required for the morphogenesis of the blastocyst.


Subject(s)
Blastocyst/physiology , Embryonic Development/physiology , Morphogenesis/physiology , Animals , Blastomeres/physiology , Cell Differentiation/physiology , Cell Division/physiology , Embryo, Mammalian/physiology , Mice
11.
Bioessays ; 28(2): 146-56, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16435292

ABSTRACT

Due to the central position of scaffold proteins in numerous signaling networks, especially in MAPK pathways, considerable efforts have been made to identify new scaffolds and to characterize their function and regulation. Most of our knowledge stems from studies of yeast MAPK scaffolds, but the identification of such scaffolds in higher eukaryotes provided a new dimension to this field and led to exciting and promising new insights into the regulation of MAPK signaling. In this review, we shortly summarize the well-established basic functions of scaffolds in yeast and highlight concepts emerging from recent studies in yeast and higher eukaryotes. In particular, we discuss how scaffolds may actively influence MAPK signaling by inducing conformational changes of bound kinases or substrates, by controlling the localization of activated MAPK and the extent and output of MAPK activation, and by modulating MAPK kinetics through the recruitment of phosphatases or ubiquitin-ligases. Finally, we summarize the current knowledge of scaffold regulation, and how these events may be functionally important for MAPK signaling.


Subject(s)
Cell Compartmentation , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Allosteric Regulation , Animals , Enzyme Activation , Substrate Specificity
12.
Dev Biol ; 271(1): 87-97, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15196952

ABSTRACT

The preimplantation development of the mouse embryo leads to the divergence of the first two cell lineages, the inner cell mass and the trophectoderm. The formation of a microvillus pole during compaction at the eight-cell stage and its asymmetric inheritance during mitosis are key events in the emergence of these two cell populations. Ezrin, a member of the ERM protein family, seems to be involved in the formation and stabilization of this apical microvillus pole. To further characterize its function in early development, we mutated the key residue T567, which was reported to be essential for regulation of ezrin function through phosphorylation. Here, we show that expression of ezrin mutants in which the COOH-terminal threonine T567 was replaced by an aspartate (to mimic a phosphorylated residue; T567D) or by an alanine (to avoid phosphorylation; T567A) interferes with E-cadherin function and disrupts the first morphogenetic events of development: compaction and cavitation. The active mutant ezrin-T567D induces the formation of numerous and abnormally long microvilli at the surface of blastomeres. Moreover, it localizes all around the cell cortex and inhibits cell-cell adhesion and cell polarization at the eight-cell stage. During the following stages, only half of the embryos are able to compact and finally to cavitate. In those embryos, the amount of ezrin-T567D decreases in the basolateral areas, while the proportion of adherens junctions increases. The reverse inactive mutant ezrin-T567A is mainly cytoplasmic and does not perturb compaction at the eight-cell stage. However, at the 16-cell stage, it relocalizes at the basolateral cortex, leading to a strong decrease in the surface of adherens junctions, and finally, embryos abort development. Our results show that ezrin is directly involved in the formation of microvilli in the early mouse embryo. Moreover, they indicate that maintenance of ezrin in basolateral areas prevents microvilli breakdown and inhibits the formation of normal cell-cell contacts mediated by E-cadherin, thereby impairing blastomeres polarization and morphogenesis of the blastocyst.


Subject(s)
Amino Acid Substitution/physiology , Gene Expression Regulation, Developmental , Mice/embryology , Microvilli/physiology , Morula/ultrastructure , Phosphoproteins/metabolism , RNA, Messenger/metabolism , Amino Acid Substitution/genetics , Animals , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Polarity/genetics , Cell Polarity/physiology , Cytoskeletal Proteins , DNA Primers , Female , Green Fluorescent Proteins , Immunohistochemistry , Luminescent Proteins , Microinjections , Microscopy, Electron , Microscopy, Video , Microvilli/genetics , Morula/physiology , Mutagenesis, Site-Directed , Phosphoproteins/genetics , Phosphoproteins/physiology , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL