首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Retinoic acid is a widely used factor in both mouse and human embryonic stem cells. It suppresses differentiation to mesoderm and enhances differentiation to ectoderm. Fibroblast growth factor 2 (FGF2) is widely used to induce differentiation to neurons in mice, yet in primates, including humans, it maintains embryonic stem cells in the undifferentiated state. In this study, we established an FGF2 low-dose-dependent embryonic stem cell line from cynomolgus monkeys and then analyzed neural differentiation in cultures supplemented with retinoic acid and FGF2. When only retinoic acid was added to culture, neurons differentiated from FGF2 low-dose-dependent embryonic stem cells. When both retinoic acid and FGF2 were added, neurons and astrocytes differentiated from the same embryonic stem cell line. Thus, retinoic acid promotes the differentiation from embryonic stem cells to neuroectoderm. Although FGF2 seems to promote self-renewal in stem cells, its effects on the differentiation of stem cells are influenced by the presence or absence of supplemental retinoic acid.Abbreviations: EB, embryoid body; ES, embryonic stem; ESM, embryonic stem cell medium; FGF, fibroblast growth factor; GFAP, glial fibrillary acidic protein; LIF, leukemia inhibitory factor; MBP, myelin basic protein; RA, retinoic acid; SSEA, stage-specific embryonic antigen; TRA, tumor-related antigenPluripotent stem cells are potential sources of material for cell replacement therapy and are useful experimental tools for in vitro models of human disease and drug screening. Embryonic stem (ES) cells are capable of extensive proliferation and multilineage differentiation, and thus ES-derived cells are suitable for use in cell-replacement therapies.18,23 Reported ES cell characteristics including tumorigenic potential, DNA methylation status, expression of imprinted genes, and chromatin structure were elucidated by using induced pluripotent stem cells.2,11,17 Because the social expectations of regeneration medicine are growing, we must perform basic research with ES cells, which differ from induced pluripotent stem cells in terms of origin, differentiation ability, and epigenetic status.2,8Several advances in research have been made by using mouse ES cells. Furthermore, primate ES cell lines have been established from rhesus monkeys (Macaca mulatta),24 common marmosets (Callithrix jacchus),25 cynomolgus monkeys (M. fascicularis),20 and African green monkeys (Chlorocebus aethiops).19 Mouse and other mammalian ES cells differ markedly in their responses to the signaling pathways that support self-renewal.8,28 Mouse ES cells require leukemia inhibitory factor (LIF)–STAT3 signaling.14 In contrast, primate ES cells do not respond to LIF. Fibroblast growth factor 2 (FGF2) appears to be the most upstream self-renewal factor in primate ES cells. FGF2 also exerts its effects through indirect mechanisms, such as the TGFβ–Activin–Nodal signaling pathway, in primate ES cells.21 In addition to the biologic similarities between monkeys and humans, ES cells derived from cynomolgus monkeys or human blastocysts have extensive similarities that are not apparent in mouse ES cells.8,14,21,28 Numerous monkey ES cell lines are now available, and cynomolgus monkeys are an efficient model for developing strategies to investigate the efficacy of ES-cell–based medical treatments in humans.Several growth factors and chemical compounds, including retinoic acid (RA),4,9,13,22,26 FGF2,9,10,16,22 epidermal growth factor,9,22 SB431542,1,4,10 dorsomorphin,10,27 sonic hedgehog,12,13,16,27,29 and noggin,1,4,9,27 are essential for the differentiation and proliferation or maintenance of neural stem cells derived from primate ES cells. Of these factors, active RA signaling suppresses a mesodermal fate by inhibiting Wnt and Nodal signaling pathways during in vitro culture and leads to neuroectoderm differentiation in ES cells.4,13,26 RA is an indispensable factor for the specialization to neural cells. FGF2 is important during nervous system development,12 and FGF2 and RA both are believed to influence the differentiation to neural cells. The current study was done to clarify the mechanism of RA and FGF2 in the induction of differentiation along the neural lineage.We recently established a monkey ES cell line that does not need FGF2 supplementation for maintenance of the undifferentiated state. This ES cell line allowed us to study the role of differentiation to neural cells with RA and enabled us to compare ES cell differentiation in the context of supplementation with RA or FGF2 in culture. To this end, we established a novel cynomolgus monkey cell line derived from ES cells and maintained it in an undifferentiated state in the absence of FGF2 supplementation.  相似文献   

2.
3.
4.
5.
Alzheimer''s disease (AD), a progressive neurodegenerative disorder that is the most common cause of dementia in the elderly, is characterized by the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles, as well as a progressive loss of synapses and neurons in the brain. The major pertinacious component of amyloid plaques is Aβ, a variably sized peptide derived from the integral membrane protein amyloid precursor protein (APP). The Aβ region of APP locates partly within its ecto- and trans-membrane domains. APP is cleaved by three proteases, designated as α-, β-, and γ-secretases. Processing by β- and γ-secretase cleaves the N- and C-terminal ends of the Aβ region, respectively, releasing Aβ, whereas α-secretase cleaves within the Aβ sequence, releasing soluble APPα (sAPPα). The γ-secretase cleaves at several adjacent sites to yield Aβ species containing 39–43 amino acid residues. Both α- and β-cleavage sites of human wild-type APP are located in APP672–699 region (ectodomain of β-C-terminal fragment, ED-β-CTF or ED-C99). Therefore, the amino acid residues within or near this region are definitely pivotal for human wild-type APP function and processing. Here, we report that one ED-C99-specific monoclonal antibody (mAbED-C99) blocks human wild-type APP endocytosis and shifts its processing from α- to β-cleavage, as evidenced by elevated accumulation of cell surface full-length APP and β-CTF together with reduced sAPPα and α-CTF levels. Moreover, mAbED-C99 enhances the interactions of APP with cholesterol. Consistently, intracerebroventricular injection of mAbED-C99 to human wild-type APP transgenic mice markedly increases membrane-associated β-CTF. All these findings suggest that APP672–699 region is critical for human wild-type APP processing and may provide new clues for the pathogenesis of sporadic AD.Abnormal functioning and/or processing of amyloid precursor protein (APP), a type I membrane protein, has a pivotal role in the pathogenesis of Alzheimer''s disease (AD).1, 2, 3 APP is cleaved by three proteases, designated as α-, β-, and γ-secretases (Supplementary Figure S1). The major fraction (>90%) of wild-type APP is proteolyzed by α-secretase that cleaves wild-type APP between residues APP687 and APP688 within the amyloid-β (Aβ) sequence, releasing soluble APPα (sAPPα) and α-C-terminal fragment (α-CTF, C83). Only a minority (<10%) of all wild-type APP molecules undergo β-cleavage at the β-cleavage site (between residues APP671 and APP672) generating sAPPβ and β-CTF (C99), the latter of which is subsequently processed by γ-secretase complex to generate a mixture of Aβ peptides primarily 40 or 42 residues in length (Aβ1-40/42).4, 5 The β-secretase cleaves APP in addition at a β′-site (between residues APP681 and APP682) to generate C89 that is further processed by γ-secretase to produce truncated Aβ11–40/42 species.6Both α- and β-cleavage sites of wild-type APP are located in APP672–699 region (the ectodomain of β-CTF, ED-β-CTF, or ED-C99; Supplementary Figure S1). Therefore, the amino acid residues within or near this region are definitely pivotal for wild-type APP function and processing. Previous studies have identified that mutation in ED-C99 region can affect the physiological processing of APP and contribute to pathological features of familial AD (fAD). For example, Swedish APP carrying APP670/671 mutation (KM→NL) is cleaved by β-secretase over 50-fold more efficiently than wild-type APP.7 APP673 mutation (A→V) and APP693 mutation (E→G) can enhance Aβ production and accelerate formation of amyloid fibrils.8, 9, 10 APP682 mutation (E→K) blocks APP β′-site and shifts cleavage to β-site, thus increasing Aβ1–40/42 production.6 Although sporadic AD (sAD), the more common type of AD comprising 90 to 95% of all AD cases, lacks mutations in the APP gene, region-specific protein modifications within the ED-C99 region may affect wild-type APP processing similarly to APP gene mutations. For example, phosphorylation of ED-C99 at the threonine 687 (of APP770 isoform, or corresponding threonine 668 of APP751 isoform; Supplementary Figure S1) facilitates APP processing by γ-secretase.11 Therefore, the elucidation of potential influences of region-specific modifications, induced by either endogenous or exogenous molecules, on wild-type APP processing would be especially critical for clarifying the mechanisms underlying the pathogenesis of sAD.To confirm this hypothesis, we used one mouse monoclonal antibody specifically recognizing ED-C99 (mAbED-C99) with its epitope at APP674–679 (Supplementary Figure S1). The influences of mAbED-C99 binding on human wild-type APP processing were evaluated in vitro using Chinese hamster ovary cells expressing human wild-type APP (CHO/APPwt cells) and cortical neurons derived from human wild-type APP transgenic (TgAPPwt) mice. The in vitro effects of ED-C99 binding with mAbED-C99 on wild-type APP processing were further evaluated and confirmed in vivo using TgAPPwt mice and 5 × FAD transgenic mice (Tg6799 line).  相似文献   

6.
7.
8.
9.
The Komeda miniature rat Ishikawa (KMI) is a spontaneous animal model of dwarfism caused by a mutation in Prkg2, which encodes cGMP-dependent protein kinase type II (cGKII). This strain has been maintained as a segregating inbred strain for the mutated allele mri. In this study, we characterized the phenotype of the KMI strain, particularly growth traits, craniofacial measurements, and organ weights. The homozygous mutant (mri/mri) animals were approximately 70% to 80% of the size of normal, heterozygous (mri/+) animals in regard to body length, weight, and naso-occipital length of the calvarium, and the retroperitoneal fat of mri/mri rats was reduced greatly. In addition, among progeny of the (BN×KMI-mri/mri)F1×KMI-mri/mri backcross, animals with the KMI phenotype (mri/mri) were easily distinguished from those showing the wild-type phenotype (mri/+) by using growth traits such as body length and weight. Genetic analysis revealed that all of the backcrossed progeny exhibiting the KMI phenotype were homozygous for the KMI allele in the 1.2-cM region between D14Rat5 and D14Rat80 on chromosome 14, suggesting strongly that mri acts in a completely recessive manner. The KMI strain is the first and only rat model with a confirmed mutation in Prkg2 and is a valuable model for studying dwarfism and longitudinal growth traits in humans and for functional studies of cGKII.Abbreviations: cGKII, cGMP-dependent protein kinase type II; CNP, C-type natriuretic peptide; KMI, Komeda miniature rat IshikawaDwarfism is caused by both endocrinologic and nonendocrinologic defects. Most instances of dwarfism, including normal variants, are nonendocrinologic, and subjects retain growth hormone secretion. Although spontaneous rodent models of dwarfism with confirmed mutations have been reported—Snell dwarf mice with Pou1f1 (Pit1) mutation,14 Ames dwarf mice with Prop1 mutation,22 little mice with Ghrhr mutation,15 pygmy mice (also known as mini-mice) with Hmga2 (HMGI-C) mutation,26 spontaneous dwarf rats with Gh mutation,23 and rdw rats with Tg mutation9,11—most of these are models of endocrinologic dwarfism. A few models of nonendocrinologic dwarfism have been produced by gene manipulation techniques, such as transgenic and knockout strategies, and include Col2a1-transgenic mice,7,24 Col10a1-transgenic mice,10 and Fgfr3-knock-in mice.13A novel spontaneous dwarf mutation, miniature rat Ishikawa (mri), was discovered in a closed colony of Wistar rats at Ishikawa Animal Laboratory (Saitama, Japan) and has been maintained on the genetic background of Wistar rats. This mutant strain, previously termed Miniature Rat Ishikawa (MRI), has recently been established as a segregating inbred strain on the Wistar genetic background, designated Komeda Miniature rat Ishikawa (KMI). The breeding record suggested that the mutation was inherited in an autosomal recessive mode. KMI rats show no abnormality in the basal amounts or distribution of several hormones, including growth hormone, luteinizing hormone, follicle-stimulating hormone, prolactin, thyroid-stimulating hormone, and adrenocorticotropic hormone, but growth hormone response to growth hormone releasing hormone is decreased.21Using positional candidate cloning of mri, we recently identified a deletion mutation in Prkg2, which encodes cGMP-dependent protein kinase type II (cGKII), and clarified a role of cGKII as a molecular switch that couples cessation of proliferation and the start of hypertrophic differentiation of chondrocytes.2 Longitudinal skeletal growth is achieved by endochondral ossification in the growth plate, in which chondrocyte hypertrophic differentiation is an important step. Due to the impaired coupling of proliferation and hypertrophic differentiation in the growth plate chondrocytes, homozygous mutant (mri/mri) animals show longitudinal growth retardation.In this study, we further characterize the phenotype of the KMI strain, including body length, body weight, organ weight, and craniofacial measurements. Furthermore, we describe phenotypic characteristics of the progeny produced from the (BN×KMI-mri/mri)F1×KMI-mri/mri backcross and provide updated genetic, physical, and comparative maps of the mri region.  相似文献   

10.
Inhibitors of phosphodiesterase 5 (PDE5) are widely used to treat erectile dysfunction and pulmonary hypertension in clinics. PDE5, cyclic guanosine monophosphate (cGMP), and protein kinase G (PKG) are important components of the non-canonical Wnt signaling. This study aimed to investigate the effect of PDE5 inhibition on canonical Wnt signaling and osteoblastogenesis, using both in vitro cell culture and in vivo animal models. In the in vitro experiments, PDE5 inhibition resulted in activation of cGMP-dependent protein kinase 2 and consequent inhibition of glycogen synthase kinase 3β phosphorylation, destabilization of cytosolic β-catenin and the ultimate suppression of canonical Wnt signaling and reduced osteoblastic differentiation in HEK293T and C3H10T1/2 cells. In animal experiments, systemic inhibition of PDE5 suppressed the activity of canonical Wnt signaling and osteoblastogenesis in bone marrow-derived stromal cells, resulting in the reduction of bone mass in wild-type adult C57B/6 mice, significantly attenuated secreted Frizzled-related protein-1 (SFRP1) deletion-induced activation of canonical Wnt signaling and excessive bone growth in adult SFRP1−/− mice. Together, these results uncover a hitherto uncharacterized role of PDE5/cGMP/PKG signaling in bone homeostasis and provide the evidence that long-term treatment with PDE5 inhibitors at a high dosage may potentially cause bone catabolism.In the canonical Wnt (Wnt/β-catenin (β-cat)) signaling cascade, Wnt binds to Frizzled (Frz) receptors and the low-density lipoprotein receptor-related protein (LRP) 5 or 6, thereby activating dishevelled, suppressing the glycogen synthase kinase 3β (GSK3β) activity and inhibiting phosphorylation of β-cat at Thr41, Ser37, and Ser33 sites. The stabilized cytosolic β-cat enters the nucleus and consequently activates its downstream target genes via lymphoid enhancer-binding factor-1 (Lef-1) and T-cell factors.1, 2 This signaling is fine-tuned in part via a negative feedback mechanism involving secreted and transmembrane Wnt inhibitors and activators, secreted Frz-related proteins (SFRPs), and Dickkopf-1 (Dkk1).3, 4Canonical Wnt signaling is critical not only to bone development in embryogenesis but also to the maintenance of bone mass during adult life.5 The initial evidence came from the discoveries that in humans loss- or gain-of-function mutations in LRP5 were linked with the osteoporosis-pseudoglioma syndrome and a high-bone-density syndrome, respectively.6, 7, 8 Subsequent studies in mice showed that Wnt signaling might promote ossification by inducing the differentiation of bone-forming osteoblasts, suppressing the development of bone-resorbing osteoclasts, and driving the differentiation of multi-potent stem cells toward an osteoblast cell fate.9Non-canonical Wnt signaling is β-cat independent and consists of two main pathways: the Rho small GTPases-mediated planar cell polarity pathway and the Wnt/Ca2+ pathway,10 involved in various aspects of cell fate differentiation and cell movement. Non-canonical Wnt signaling has profound effects on tissue morphogenesis in a variety of vertebrate species.10 The potential role for non-canonical Wnt signaling in bone formation has been investigated recently in limited studies, which have shown that the non-canonical Wnt-Gαq/11-PKC pathway operates in mammalian osteoprogenitors to promote osteoblast development, and that Wnt16 exhibits a stimulatory effect on bone metabolism.11, 12, 13 Nevertheless, the molecular events in the non-canonical Wnt signaling regulation of bone development and homeostasis have yet to be further elucidated.Phosphodiesterases (PDEs) are a large family of enzymes that cleave cyclic nucleotides. To date, 11 PDE subtypes have been identified, among which PDE5 has been most extensively studied. PDE5, cyclic guanosine monophosphate (cGMP), and cGMP-dependent protein kinase (PKG) are among the major components of the non-canonical Wnt signaling pathway and are involved in the regulation of intracellular Ca2+ concentration.14, 15 It is now well established that PDE5 degrades 3''-5′- cGMP and its inhibition leads to an increase in intracellular cGMP levels and activation of protein kinase G (PKG), resulting in a decrease in Ca2+ influx and consequent relaxation of smooth muscles, which produces the therapeutic effects in clinical erectile dysfunction (ED) and pulmonary hypertension (PH).14 Currently, little is known regarding the involvement of PDE5 in Wnt signaling regulation of bone formation and homeostasis. The objective of this study was to determine the effect of PDE5 inhibition on canonical Wnt signaling and bone mass.  相似文献   

11.
12.
13.
14.
Although the mechanisms controlling skeletal muscle homeostasis have been identified, there is a lack of knowledge of the integrated dynamic processes occurring during myogenesis and their regulation. Here, metabolism, autophagy and differentiation were concomitantly analyzed in mouse muscle satellite cell (MSC)-derived myoblasts and their cross-talk addressed by drug and genetic manipulation. We show that increased mitochondrial biogenesis and activation of mammalian target of rapamycin complex 1 inactivation-independent basal autophagy characterize the conversion of myoblasts into myotubes. Notably, inhibition of autophagic flux halts cell fusion in the latest stages of differentiation and, conversely, when the fusion step of myocytes is impaired the biogenesis of autophagosomes is also impaired. By using myoblasts derived from p53 null mice, we show that in the absence of p53 glycolysis prevails and mitochondrial biogenesis is strongly impaired. P53 null myoblasts show defective terminal differentiation and attenuated basal autophagy when switched into differentiating culture conditions. In conclusion, we demonstrate that basal autophagy contributes to a correct execution of myogenesis and that physiological p53 activity is required for muscle homeostasis by regulating metabolism and by affecting autophagy and differentiation.Muscle satellite cells (MSCs) in adult muscle remain quiescent until external stimuli (such as injury or even exercise) trigger their re-entry into the cell cycle. Their progeny, myoblasts, fuse to form new multinucleated myofibers. In this study, the ability of MSC to give rise to muscle progenitor cells (that is, myoblasts) that could differentiate and fuse in vitro has been exploited to analyze the integrated network of signaling pathways that operate during myogenesis.Autophagy undergoes a fine tuning during cell and tissue differentiation in order to adapt to the dynamic changes occurring in the tissue microenvironment.1 Using the stable C2C12 cell line, it was shown that autophagy is induced during muscle differentiation despite the concomitant activation of mammalian target of rapamycin (mTOR).2 Interestingly, inhibition of autophagy was found to impair the differentiation and fusion of C2C12 myoblasts, while favoring their apoptosis.3 Autophagy is increased in muscle in several physiological and pathological conditions, including fasting, atrophy and exercise.4 Much less is known about the link between cell metabolism and autophagy during muscle differentiation under physiological conditions.p53 has been shown to promote myoblast differentiation by regulating the function of pRb,5, 6 and to have a pleiotropic role in muscle metabolism by promoting exercise-induced mitochondrial biogenesis in skeletal muscle.7, 8 How the physiological level of p53 impacts on these changes during differentiation has not been explored.The role of p53 in the regulation of autophagy is multi-facets.9 Nuclear p53 positively regulates autophagy following exogenous stress, resulting in a pro-death or pro-survival outcomes.10 Conversely, cytoplasmic p53 inhibits autophagy under starvation or endoplasmic reticulum (ER) stress.11 What is the role of p53 in the regulation of basal autophagy during myogenesis and its physiological implications are still unknown.We address these issues using mouse skeletal MSC-derived myoblasts that when differentiate in vitro well mimic the dynamic processes occurring in vivo when a myoblast is asked to differentiate and fuse into a fully differentiated myotube. The findings here reported unravel a clear role for basal autophagy in muscle differentiation and identify a role for p53 in muscle metabolism and basal autophagy.  相似文献   

15.
16.
17.
18.
Both fibroblast growth factor 9 (Fgf9) and Kit Ligand (Kl) signal through tyrosine kinase receptors, yet they exert opposite effects on meiotic differentiation in postnatal spermatogonia, Fgf9 acting as a meiosis-inhibiting substance and Kl acting as a promoter of the differentiation process. To understand the molecular mechanisms that might underlie this difference, we tried to dissect the intracellular signaling elicited by these two growth factors. We found that both Fgf9 and Kl stimulate Erk1/2 activation in Kit+ (differentiating) spermatogonia, even though with different time courses, whereas Kl, but not Fgf9, elicits activation of the Pi3k-Akt pathway. Sustained Erk1/2 activity promoted by Fgf9 is required for induction of the autocrine Cripto-Nodal-Smad2/3 signaling loop in these cells. Nodal signaling, in turn, is essential to mediate Fgf9 suppression of the meiotic program, including inhibition of Stra8 and Scp3 expression and induction of the meiotic gatekeeper Nanos2. On the contrary, sustained activation of the Pi3k-Akt pathway is required for the induction of Stra8 expression elicited by Kl and retinoic acid. Moreover, we found that Kl treatment impairs Nodal mRNA expression and Fgf9-mediated Nanos2 induction, reinforcing the antagonistic effect of these two growth factors on the meiotic fate of male germ cells.In the mouse testis, competence to enter meiosis is acquired during the differentiative stages in which spermatogonia undergo Kit-dependent mitotic divisions, but not in Kit-negative spermatogonial stem cells. Retinoic acid (RA) stimulates Kit expression in spermatogonia and Kit Ligand (Kl) expression in Sertoli cells.1 Kl is essential to promote the mitotic expansion of Kit+ premeiotic germ cells.2 However, the concerted action of both RA and Kl can induce meiotic entry of in vitro cultured Kit+ spermatogonia.1 Indeed, both RA and Kl increase the expression of genes that are fundamental for the beginning of the meiotic process,1, 3 and in particular of Stimulated by Retinoic Acid Gene 8 (Stra8), which is essential for the switch from the mitotic cell cycle to the meiotic program.1 Selective inhibitors of Kit tyrosine-kinase activity block both RA- and Kl-induced meiotic entry, suggesting that the two factors converge on common, Kit-dependent, signaling pathways.1 RA- and Kl-induced meiotic entry is mediated, at least in part, by activation of the Pi3k-Akt pathway. RA promotes meiosis also by inhibiting the expression of the meiotic gatekeeper Nanos2, an RNA-binding protein that silences genes essential for spermatogonial differentiation and meiotic entry.4 Fibroblast growth factor 9 (Fgf9) secreted by Sertoli cells acts as a meiosis-inhibiting substance by increasing Nanos2 levels in premeiotic spermatogonia, thus opposing the RA/Kl/Kit axis.4 In the male fetal gonad, meiotic entry of male fetal gonocytes is inhibited by the action of Cyp26b1, which degrades RA of mesonephric origin.5, 6 Fgf9 contributes to prevent meiosis in the male fetal gonad by inducing Nanos2 expression in gonocytes.4, 7 Several recent works have shown that Nodal (a Tgfβ superfamily member) plays an important role in the inhibition of the meiotic program of fetal male germ cells.8, 9, 10 Moreover, it has been demonstrated that the Fgf9 inhibitory effect on meiotic entry of fetal gonocytes might be mediated, at least in part, by activation of Cripto-Nodal signaling.9In the present work, we show that the Fgf9 antimeiotic effect is mediated by activation of the Cripto-Nodal-Smad2/3 signaling in postnatal spermatogonia, and that this activation requires Fgf9-dependent stimulation of the Erk1/2 pathway. On the contrary, Kl promotes meiotic differentiation through the activation of the Pi3k-Akt pathway and inhibition of Nodal expression.  相似文献   

19.
20.
Tissue-type plasminogen activator (tPA) is a pleiotropic serine protease of the central nervous system (CNS) with reported neurotrophic and neurotoxic functions. Produced and released under its single chain form (sc), the sc-tPA can be cleaved by plasmin or kallikrein in a two chain form, tc-tPA. Although both sc-tPA and tc-tPA display a similar fibrinolytic activity, we postulated here that these two conformations of tPA (sc-tPA and tc-tPA) could differentially control the effects of tPA on neuronal survival. Using primary cultures of mouse cortical neurons, our present study reveals that sc-tPA is the only one capable to promote N-methyl-D-aspartate receptor (NMDAR)-induced calcium influx and subsequent excitotoxicity. In contrast, both sc-tPA and tc-tPA are capable to activate epidermal growth factor receptors (EGFRs), a mechanism mediating the antiapoptotic effects of tPA. Interestingly, we revealed a tPA dependent crosstalk between EGFR and NMDAR in which a tPA-dependent activation of EGFRs leads to downregulation of NMDAR signaling and to subsequent neurotrophic effects.Tissue-type plasminogen activator (tPA) is secreted by endothelial cells and promotes fibrinolysis via the conversion of fibrin-bound plasminogen into plasmin.1 Neurons and some glial cells also secrete tPA.2, 3, 4, 5 tPA is secreted as a single-chain form (sc-tPA), which can be processed into a two-chain form (tc-tPA) by plasmin or kallikreins.6, 7 Interestingly, sc-tPA is proteolytically active even without proteolytic processing. In addition to its vascular functions, tPA displays critical roles in the brain parenchyma with roles in cell migration, neuronal plasticity and survival,8, 9, 10, 11, 12, 13, 14 acting either as an enzyme or as a cytokine-like molecule. Among its actions, tPA is well described to promote neurotoxicity, likely through promotion of N-methyl-D-aspartate receptor (NMDAR) activity.15, 16, 17 Recently, we reported that only sc-tPA can promote NMDAR signaling and neurotoxicity.18 Interestingly, data from wild-type mice,19 transgenic mice overexpressing tPA in neurons20 or in vitro21 also report neuroprotective effects of tPA.9, 10 The proposed mechanisms involved a tPA-dependent and non-proteolytic activation of either epidermal growth factor receptors (EGFRs)22 on oligodendrocytes or NMDARs.20Here we explored a link between tPA conformations (sc-tPA and tc-tPA), EGFR- and NMDAR-dependent signaling pathways. Our findings identify sc-tPA as a selective positive modulator of NMDAR signaling in neurons when present at high concentrations and both sc-tPA and tc-tPA as positive modulators of EGFR signaling, this even at low concentrations. We also reveal a crosstalk between these two families of receptors, with the tPA-dependent activation of EGFRs reducing NMDAR signaling. By these mechanisms, sc-tPA and tc-tPA control neuronal death and survival.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号