• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 14
  • 4
  • 2
  • 1
  • Tagged with
  • 22
  • 9
  • 9
  • 9
  • 8
  • 8
  • 8
  • 8
  • 7
  • 7
  • 7
  • 6
  • 5
  • 5
  • 5
  • 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.
1

Role of Nestin in Mouse Development

Mohseni, Paria 05 March 2012 (has links)
Although nestin has served as a marker of neural stem/progenitor cells for close to twenty years, its function is still poorly understood. During development, this intermediate filament protein is expressed in many different progenitors including those of the central nervous system, heart, skeletal muscle and kidney. The adult expression of nestin is mainly restricted to the subependymal zone and dentate gyrus of the brain, the neuromuscular junction and renal podocytes. I have used two approaches of gain of function and loss of function to elucidate the role of nestin in vivo. Although I was able to generate transgenic lines in which the transgene was ubiquitously expressed at the RNA level, over-expression of nestin at the protein level was not achieved possibly due to post transcriptional regulation of this gene. My data from loss of function approach indicates that nestin-deficient mice have impaired coordination. Balance and muscle strength are not affected and there are no apparent anatomical defects. I found that nestin deficiency is compatible with normal development of the central nervous system but results in abnormal clustering of acetylcholine receptors in the neuromuscular junctions, similar to the phenotype described for deficiency of cyclin-dependent kinase 5 (Cdk5) a candidate downstream effector of nestin. In renal podocytes, where both nestin and Cdk5 are normally expressed, we found reduced branching and abnormally contoured podocyte processes. To further connect the phenotype of nestin deficiency to Cdk5, I demonstrated that nestin deficiency can rescue maintenance of acetylcholine receptor clusters in the absence of agrin, similar to Cdk5/agrin double knockouts, indicating that the observed nestin deficiency phenotypes are the consequence of aberrant Cdk5 activity.
2

Role of Nestin in Mouse Development

Mohseni, Paria 05 March 2012 (has links)
Although nestin has served as a marker of neural stem/progenitor cells for close to twenty years, its function is still poorly understood. During development, this intermediate filament protein is expressed in many different progenitors including those of the central nervous system, heart, skeletal muscle and kidney. The adult expression of nestin is mainly restricted to the subependymal zone and dentate gyrus of the brain, the neuromuscular junction and renal podocytes. I have used two approaches of gain of function and loss of function to elucidate the role of nestin in vivo. Although I was able to generate transgenic lines in which the transgene was ubiquitously expressed at the RNA level, over-expression of nestin at the protein level was not achieved possibly due to post transcriptional regulation of this gene. My data from loss of function approach indicates that nestin-deficient mice have impaired coordination. Balance and muscle strength are not affected and there are no apparent anatomical defects. I found that nestin deficiency is compatible with normal development of the central nervous system but results in abnormal clustering of acetylcholine receptors in the neuromuscular junctions, similar to the phenotype described for deficiency of cyclin-dependent kinase 5 (Cdk5) a candidate downstream effector of nestin. In renal podocytes, where both nestin and Cdk5 are normally expressed, we found reduced branching and abnormally contoured podocyte processes. To further connect the phenotype of nestin deficiency to Cdk5, I demonstrated that nestin deficiency can rescue maintenance of acetylcholine receptor clusters in the absence of agrin, similar to Cdk5/agrin double knockouts, indicating that the observed nestin deficiency phenotypes are the consequence of aberrant Cdk5 activity.
3

Characterization of Nestin Proteins in the Goldfish: Implications for Regeneration of Adult Dopaminergic Neurons

Venables, Maddie Jolyane January 2016 (has links)
Nestin is a type VI intermediate filament protein that marks proliferative cells in the central and peripheral nervous system of vertebrates during development and adulthood. Nestin is not only expressed in progenitor cells of neuronal tissues but is also present in muscle, heart, lung, pancreas and skin follicle tissues. The goal of this thesis is to investigate and characterize the nestin protein in goldfish and relate nestin expression to neuroregeneration and brain plasticity events in the adult goldfish forebrain. Currently little is known about nestin function and regulation in vertebrates, especially in fish. In this study we used Rapid amplification of cDNA ends PCR (RACE-PCR) to isolate goldfish nestin mRNA. We uncovered several different mRNA transcripts. PCR analysis and sequencing further identified three different nestin transcripts of 4003, 2446, and 2126 nucleotides with a predicted protein length of 860, 274, and 344 amino acids respectively. We next applied a multiple-antigenic peptide (MAP) strategy to generate a polyclonal goldfish-specific nestin antibody against a 23 amino acid sequence located at the N-terminal end of goldfish nestin. Western blotting revealed the existence of three different nestin protein isoforms (nestin A, B and C); the first report of nestin isoforms in teleost species. Nestin expression and distribution in the goldfish brain is complex and revealed both individual and tissue-dependent variations. The most remarkable finding following principal component analysis of the western blot data was the uniqueness of the pituitary, hypothalamus and telencephalon. These tissues are proliferative in nature containing progenitor and proliferative cellular pools that are involved in important biological axes such as the motor and reproductive axis. Interestingly, all three tissues were able to change their proliferative cellular profile of nestin protein expression to alleviate the detrimental effects of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) upon administration. The toxin MPTP destroys dopamine neurons in the fish brain leading to motor deficits and reproductive difficulties. The incorporation of 5-bromo-2’-deoxyuriding (BrdU) into newly synthesized DNA revealed an upregulation of BrdU immunolabeling following MPTP administration in the area telencephali pars dorsalis (Vd) and along the ventricular surface area of the telencephalon suggesting the generation of new neurons in the adult central nervous system. This thesis reports novel nestin isoforms and illustrates regenerative events occurring in the goldfish telencephalon following a neurotoxic insult. This work provides a framework for future investigations of the differential roles and regulation of the nestins to better understand seasonal neuronal plasticity, neuronal regeneration and neuronal circuitry in teleost.
4

The Stem Cell Marker Nestin is Critical for TGF beta1- Mediated Tumor Progression in Pancreatic Cancer

Su, Huei-Ting 25 June 2012 (has links)
Stem cell marker Nestin is an intermediate filament protein that plays an important role in cell integrity, migration and differentiation. Nestin expression occurs in approximately one-third of pancreatic ductal adenocarcinoma (PDAC) cases, and its expression positively correlates with tumor stage and peripancreatic invasion. Little is known of the mechanisms by which Nestin influences PDAC progression. We showed that Nestin overexpression in PDAC cells increased cell motility and drove phenotypic changes associated with the epithelial-mesenchymal transition in vitro, conversely, knockdown of endogenous Nestin expression reduced the migration rate and cells reverted to a more epithelial phenotype. In vivo mice studies showed that knockdown of Nestin significantly reduced tumor incidence and volume in xenografts. Expression of the Nestin protein was associated with Smad4 status in PDAC cells, hence Nestin expression might be regulated by the TGF-b1/SMAD4 pathway in PDAC. We examined Nestin expression after TGF-b1 treatment in human pancreatic cancer PANC-1, and PANC-1 shSmad4 cells. The TGF-b/SMAD pathway induced Nestin protein expression in PDAC cells through Smad4 in a dependent manner. Moreover, increased Nestin expression caused a positive feedback loop in the TGFb/SMAD signaling system. Finally, we demonstrated that 2 anti-microtubule inhibitors, Cytochalasin D (CD) and Withaferin A (WFA), exhibited anti-Nestin activity; these inhibitors might be potential anti-metastatic drugs. Our findings uncovered a novel role of Nestin in regulating TGF-b1-induced EMT. Anti-Nestin therapeutics are under development as a potential treatment for PDAC metastasis.
5

The role of interleukin-1beta on the expression of nestin in cardiac neural stem cells following myocardial infarction

Khan, Alexandre 12 1900 (has links)
Le mécanisme biologique responsable pour l’augmentation de l’expression de la protéine nestin dans les cellules souches neurales (CSN) du cœur après un infarctus du myocarde (IM) demeure inconnu. Des études antérieures ont démontré que le traitement au dexamethasone, un glucocorticoïde aux propriétés anti-inflammatoires, abolit la régulation positive de nestin après un IM. Ceci suggère un lien avec la réponse inflammatoire. Nous avons vérifié dans cette étude l’hypothèse que la cytokine inflammatoire interleukin-1beta (IL-1beta) peut modifier le phénotype de cellules souches neurales. Le deuxième objectif de l’étude fut d’établir l’impact, suivant un IM, de l’inhibition de la signalisation de IL-1beta sur la fonction et la guérison cardiaque. Suite à une ligature complète de l’artère coronaire du rat mâle, le dysfonctionnement contractile du ventricule gauche fut associé à une régulation positive de la protéine nestin dans le myocarde non-infarci. Le traitement avec Xoma 052 (1 mg/kg), un anticorps anti-IL-1beta, 24h, 7 et 14 jours après un évènement ischémique, eu aucun effet sur la taille de l’infarctus ou la contractilité du ventricule gauche. De plus, le traitement avec Xoma 052 après un IM n’a pu supprimer l’augmentation de l’expression de nestin et Bcl-2 malgré une réduction modeste du niveau de la protéine Bax. Pour déterminer directement le rôle de la réponse inflammatoire en l’absence d’ischémie, nous avons injecté des rats mâles avec du LPS (10mg/kg, 18hrs). Dans le coeur du rat-LPS, nous avons noté une augmentation significative du niveau d’ARNm de IL-1beta et de l’expression de la protéine nestin. Le prétraitement avec 10mg/kg de Xoma 052 a aboli l’augmentation de l’expression de nestin dans le coeur des rats-LPS. Ces observations indiquent que les cellules souches neurales pourraient représenter une cible potentielle de l’IL-1beta. / The underlying biological event(s) implicated in the increased expression of nestin in cardiac resident neural stem cells (NSC) following myocardial infarction (MI) remain unknown. Past studies have shown that dexamethasone treatment, a synthetic glucocorticoid with potent anti-inflammatory properties, abolished the upregulation of nestin in the non-infacted left ventricle (NILV) following MI. This suggests an association with the inflammatory response. The present study tested the hypothesis that the inflammatory cytokine interleukin-1beta (IL-1beta) can influence the neural stem cell phenotype. A secondary goal of the study was to assess impact of IL-1beta signalling inhibition on cardiac function and wound healing following MI. Following complete coronary artery ligation of the adult male rat heart, left ventricular contractile dysfunction was associated with the upregulation of the pro-apoptotic protein bax, the anti-apoptotic protein Bcl-2 and nestin in the non-infarcted myocardium. Xoma 052 (1 mg/kg), an IgG antibody directed against IL-1beta, was administred 24 hrs after ischemic injury and subsequently injected 2x over a period of 21 days. Treatment did not alter infarct size or improve left ventricular contractility. In addition, it failed to abolish the increased expression of nestin and Bcl-2, and modestly reduced bax protein levels. To directly assess the relationship between inflammation and the expression of nestin in the absence of an ischemic insult, adult male rats were injected with lipopolysaccharide (LPS) (10mg/kg;18 hrs). In the heart of LPS- treated rats, IL-1beta mRNA levels were significantly increased and associated with elevated nestin protein expression. The pre-treatment with Xoma 052 (10mg/kg) abolished the increased expression of nestin in the heart of LPS-treated rats. These data indicate that neural stem cells may represent a target for IL-1beta. However, futher investigation is required to elucidate the role of IL-1beta on NSC following MI.
6

Regulation der Nestinexpression bei der epithelial-mesenchymalen Transition / Regulation of nestin expression in the epithelial-mesenchymal transition

Lotzkat, Anja Franziska 28 February 2012 (has links)
No description available.
7

Identification, regulation and lineage tracing of embryonic olfactory progenitors

Murdoch, Barbara 11 1900 (has links)
Neurogenesis occurs in exclusive regions in the adult nervous system, the subventricular zone and dentate gyrus in the brain, and olfactory epithelium (OE) in the periphery. Cell replacement after death or injury, occurs to varying degrees in neural tissue, and is thought to be dependent upon the biological responses of stem and/or progenitor cells. Despite the progress made to identify adult OE and central nervous system (CNS) progenitors and lineage trace their progeny, our spatial and temporal understanding of embryonic OE neuroglial progenitors has been stalled by the paucity of identifiable genes able to distinguish individual candidate progenitors. In the developing CNS, radial glia serve as both neural progenitors and scaffolding for migrating neuroblasts and are identified by the expression of a select group of antigens, including nestin. Here, I show that the embryonic OE contains a novel radial glial-like progenitor (RGLP) that is not detected in adult OE. RGLPs express the radial glial antigens nestin, GLAST and RC2, but not brain lipid binding protein (BLBP), which, distinct from CNS radial glia, is instead found in olfactory ensheathing cells, a result confirmed using lineage tracing with BLBP-cre mice. Nestin-cre-mediated lineage tracing with three different reporters reveals that only a subpopulation of nestin-expressing RGLPs activate the “CNS-specific” nestin regulatory elements, and produce spatially restricted neurons in the OE and vomeronasal organ. The dorsal-medial restriction of transgene-activating cells is also seen in the embryonic OE of Nestin-GFP transgenic mice, where GFP is found in a subpopulation of GFP+ Mash1+ neuronal progenitors, despite the fact that endogenous nestin expression is found in RGLPs throughout the OE. In vitro, embryonic OE progenitors produce three biologically distinct colony subtypes, that when generated from Nestin-cre/ZEG mice, produce GFP+ neurons, recapitulating their in vivo phenotype, and are enriched for the most neurogenic colony subtype. Neurogenesis in vitro is driven by the proliferation of nestin+ progenitors in response to FGF2. I thus provide evidence for a novel neurogenic precursor, the RGLP of the OE, that can be regulated by FGF2, and provide the first evidence for intrinsic differences in the origin and spatiotemporal potential of distinct progenitors during OE development.
8

The muscle cytoskeleton of mice and men : Structural remodelling in desmin myopathies

Carlsson, Lena January 2001 (has links)
The muscle fibre cytoskeleton of skeletal and heart muscle cells is composed mainly of intermediate filaments (IFs), that surround the myofibrils and connect the peripheral myofibrils with the sarcolemma and the nuclear membrane. Desmin is the first muscle specific IF protein to be produced in developing muscles and is the main IF protein in mature muscles. In skeletal muscle, desmin is particularly abundant at myotendinous and neuromuscular junctions. In the heart an increased amount of desmin is found at intercalated discs and in Purkinje fibres of the conduction system. Interactions between the IFs themselves, and between IFs and other structures such as Z-discs and the sarcolemma, are mediated by intermediate filament associated proteins (IFAPs). A transgenic mice model, which lacks the desmin gene have been developed to study the function of desmin. In these mice, morphological abnormalities are observed in both heart and skeletal muscles. Similar defects have been observed in human myopathies, caused by different mutations in the desmin gene. In the present thesis, skeletal and heart muscles of both wild type and desmin knock-out (K/O) mice have been investigated. Furthermore the cytoskeletal organisation in skeletal muscles from human controls and from a patient with desmin myopathy was examined. In the desmin K/O mice, no morphological alterations were observed during embryogenesis. These mice postnatally developed a cardiomyopathy and a muscle dystrophy in highly used skeletal muscles. Ruptures of the sarcolemma appear to be the primary event leading to muscle degeneration and fibrosis both in cardiac and affected skeletal muscles. In the heart the muscle degeneration gave rise to calcifications, whereas in skeletal muscles regeneration of affected muscle was seen. In mature wild type mice, the IF proteins synemin and paranemin, and the IFAP plectin were present together with desmin at the myofibrillar Z-discs, the sarcolemma, the neuromuscular junctions and the myotendinous junctions. Nestin was only found in these junctional regions. In desmin K/O mice, all four proteins were detected at neuromuscular and myotendinous junctions. The normal network of synemin and paranemin were not observed, whereas the distribution of plectin was preserved. In normal human muscles, synemin, paranemin, plectin and αB-crystallin were colocalised with desmin in between the myofibrils, at the sarcolemma and at myotendinous and neuromuscular junctions. In the human desmin myopathy, the distribution of desmin varied considerably. A normal pattern was seen in some fibres areas, whereas other regions either contained large subsarcolemmal and intermyofibrillar accumulations of desmin or totally lacked desmin. Nestin, synemin, paranemin, plectin and αB-crystallin also exhibited an abnormal distribution. They were often aggregated in the areas that contained accumulations of desmin. In cultured satellite cells from the patient, a normal network of desmin was present in early passages, whereas aggragates of desmin occurred upon further culturing. In the latter, also the nestin network was disrupted, whereas vimentin showed a normal pattern. αB-crystallin was only present in cells with a disrupted desmin network. Plectin was present in a subset of cells, irrespective of whether desmin was aggregated or showed a normal network. From the present study it can be concluded that an intact desmin network is needed to maintain the integrity of muscle fibres. Desmin may be an important component in the assembly of proteins, which connect the extrasarcomeric cytoskeleton with the extracellular matrix.
9

Identification, regulation and lineage tracing of embryonic olfactory progenitors

Murdoch, Barbara 11 1900 (has links)
Neurogenesis occurs in exclusive regions in the adult nervous system, the subventricular zone and dentate gyrus in the brain, and olfactory epithelium (OE) in the periphery. Cell replacement after death or injury, occurs to varying degrees in neural tissue, and is thought to be dependent upon the biological responses of stem and/or progenitor cells. Despite the progress made to identify adult OE and central nervous system (CNS) progenitors and lineage trace their progeny, our spatial and temporal understanding of embryonic OE neuroglial progenitors has been stalled by the paucity of identifiable genes able to distinguish individual candidate progenitors. In the developing CNS, radial glia serve as both neural progenitors and scaffolding for migrating neuroblasts and are identified by the expression of a select group of antigens, including nestin. Here, I show that the embryonic OE contains a novel radial glial-like progenitor (RGLP) that is not detected in adult OE. RGLPs express the radial glial antigens nestin, GLAST and RC2, but not brain lipid binding protein (BLBP), which, distinct from CNS radial glia, is instead found in olfactory ensheathing cells, a result confirmed using lineage tracing with BLBP-cre mice. Nestin-cre-mediated lineage tracing with three different reporters reveals that only a subpopulation of nestin-expressing RGLPs activate the “CNS-specific” nestin regulatory elements, and produce spatially restricted neurons in the OE and vomeronasal organ. The dorsal-medial restriction of transgene-activating cells is also seen in the embryonic OE of Nestin-GFP transgenic mice, where GFP is found in a subpopulation of GFP+ Mash1+ neuronal progenitors, despite the fact that endogenous nestin expression is found in RGLPs throughout the OE. In vitro, embryonic OE progenitors produce three biologically distinct colony subtypes, that when generated from Nestin-cre/ZEG mice, produce GFP+ neurons, recapitulating their in vivo phenotype, and are enriched for the most neurogenic colony subtype. Neurogenesis in vitro is driven by the proliferation of nestin+ progenitors in response to FGF2. I thus provide evidence for a novel neurogenic precursor, the RGLP of the OE, that can be regulated by FGF2, and provide the first evidence for intrinsic differences in the origin and spatiotemporal potential of distinct progenitors during OE development.
10

Identification, regulation and lineage tracing of embryonic olfactory progenitors

Murdoch, Barbara 11 1900 (has links)
Neurogenesis occurs in exclusive regions in the adult nervous system, the subventricular zone and dentate gyrus in the brain, and olfactory epithelium (OE) in the periphery. Cell replacement after death or injury, occurs to varying degrees in neural tissue, and is thought to be dependent upon the biological responses of stem and/or progenitor cells. Despite the progress made to identify adult OE and central nervous system (CNS) progenitors and lineage trace their progeny, our spatial and temporal understanding of embryonic OE neuroglial progenitors has been stalled by the paucity of identifiable genes able to distinguish individual candidate progenitors. In the developing CNS, radial glia serve as both neural progenitors and scaffolding for migrating neuroblasts and are identified by the expression of a select group of antigens, including nestin. Here, I show that the embryonic OE contains a novel radial glial-like progenitor (RGLP) that is not detected in adult OE. RGLPs express the radial glial antigens nestin, GLAST and RC2, but not brain lipid binding protein (BLBP), which, distinct from CNS radial glia, is instead found in olfactory ensheathing cells, a result confirmed using lineage tracing with BLBP-cre mice. Nestin-cre-mediated lineage tracing with three different reporters reveals that only a subpopulation of nestin-expressing RGLPs activate the “CNS-specific” nestin regulatory elements, and produce spatially restricted neurons in the OE and vomeronasal organ. The dorsal-medial restriction of transgene-activating cells is also seen in the embryonic OE of Nestin-GFP transgenic mice, where GFP is found in a subpopulation of GFP+ Mash1+ neuronal progenitors, despite the fact that endogenous nestin expression is found in RGLPs throughout the OE. In vitro, embryonic OE progenitors produce three biologically distinct colony subtypes, that when generated from Nestin-cre/ZEG mice, produce GFP+ neurons, recapitulating their in vivo phenotype, and are enriched for the most neurogenic colony subtype. Neurogenesis in vitro is driven by the proliferation of nestin+ progenitors in response to FGF2. I thus provide evidence for a novel neurogenic precursor, the RGLP of the OE, that can be regulated by FGF2, and provide the first evidence for intrinsic differences in the origin and spatiotemporal potential of distinct progenitors during OE development. / Medicine, Faculty of / Medicine, Department of / Experimental Medicine, Division of / Graduate

Page generated in 0.0465 seconds