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.
Despite enormous efforts, the patterns of the rise of eukaryotic life on Earth are not clearly defined. The ability of eukaryotes to produce energy using oxygen and sugars was a key factor in advancing life on Earth towards complex multicellular organisms. However, this was not the only way to produce energy and survive. Mitochondria probably appeared soon after the oxygen increase in the Earth's atmosphere but many microaerophilic protists require little or no oxygen to survive. New ultrastructural, biochemical and molecular phylogeny data about structures and processes involved in the generation of energy by currently known protists have forced the revision of understanding of the "tree of life".  相似文献   

2.
The cytoplasmic membrane protein FeoB of Escherichia coli, Helicobacter pylori, Legionella pneumophila and Synechocystis sp. strain PCC 6803 is necessary for Fe(2+) uptake. The C-terminal part of FeoB is predicted to contain 8-12 membrane-spanning helices. The N-terminal domain shows much similarity to eukaryotic and prokaryotic G proteins and, indeed, GTPase activity is necessary for Fe(2+) transport. Four of the five characteristic conserved G protein motifs have been identified in FeoB proteins. Whether FeoB is involved directly, via its Me(2+) binding site, or indirectly in Fe(2+) transport, remains to be investigated.  相似文献   

3.
Is loss of function of the prion protein the cause of prion disorders?   总被引:4,自引:0,他引:4  
Transmissible spongiform encephalopathies are fatal neurodegenerative diseases that involve misfolding of the prion protein. Recent studies have provided evidence that normal prion protein might have a physiological function in neuroprotective signaling, suggesting that loss of prion protein activity might contribute to the pathogenesis of prion disease. However, studies using knockout animals do not support the loss-of-function hypothesis and argue that prion neurodegeneration might be associated with a gain of a toxic activity by the misfolded prion protein. Thus, the mechanism of neurodegeneration in spongiform encephalopathies remains enigmatic.  相似文献   

4.
《朊病毒》2013,7(4):202-205
Tens of putative interacting partners of the cellular prion protein (PrPC) have been identified, yet the physiologic role of PrPC remains unclear. For the first time, however, a recent paper has demonstrated that the absence of PrPC produces a lethal phenotype. Starting from this evidence, here we discuss the validity of past and more recent literature supporting that, as part of protein platforms at the cell surface, PrPC may bridge extracellular matrix molecules and membrane proteins to intracellular signaling pathways.  相似文献   

5.
Is green fluorescent protein toxic to the living cells?   总被引:27,自引:0,他引:27  
Green fluorescent protein (GFP) has become more popular to be used as a living marker for positively transfected clones in many studies. To establish stable cell lines constitutively expressing GFP, three GFPs expressed from plasmid pBIEGFP, pSG5GFP, and pRSGFP were introduced into NIH/3T3, BHK-21, Huh-7, and HepG2 cells. All the GFPs we used are the mutant forms of a common wild phenotype. The pBIEGFP expressed enhanced GFP (EGFP). The pRSGFP and pSG5GFP expressed red shift GFP (RSGFP). The RSGFP gene in pSG5GFP was driven by a strong SV40 promoter and showed at least 20-fold higher RSGFP expression by western blot analysis. Despite of the variation in the levels of GFP expression, many GFP expressing cells contracted, rounded-up, and died, which was confirmed by decreasing luciferase activity. CPP32 activity and flow cytometric analyses further demonstrate that cells expressing GFP underwent apoptosis. Our observation is contradictory to other reports that GFP is nontoxic to the cells. Most importantly, this paper shows for the first time the link between expression of GFP and induction of apoptosis. This finding should promote studies of GFP cytotoxicity and attempts to isolate new non-toxic mutants of GFP.  相似文献   

6.
The native prion protein (PrP) has a two domain structure, with a globular folded α-helical C-terminal domain and a flexible extended N-terminal region. The latter can selectively bind Cu2+ via four His residues in the octarepeat (OR) region, as well as two sites (His96 and His111) outside this region. In the disease state, the folded C-terminal domain of PrP undergoes a conformational change, forming amorphous aggregates high in β-sheet content. Cu2+ bound to the ORs can be redox active and has been shown to induce cleavage within the OR region, a process requiring conserved Trp residues. Using computational modeling, we have observed that electron transfer from Trp residues to copper can be favorable. These models also reveal that an indole-based radical cation or Cu+ can initiate reactions leading to protein backbone cleavage. We have also demonstrated, by molecular dynamics simulations, that Cu2+ binding to the His96 and His111 residues in the remaining PrP N-terminal fragment can induce localized β-sheet structure, allowing us to suggest a potential mechanism for the initiation of β-sheet misfolding in the C-terminal domain by Cu2+.
Hans J. VogelEmail:
  相似文献   

7.
Tens of putative interacting partners of the cellular prion protein (PrPC) have been identified, yet the physiologic role of PrPC remains unclear. For the first time, however, a recent paper has demonstrated that the absence of PrPC produces a lethal phenotype. Starting from this evidence, here we discuss the validity of past and more recent literature supporting that, as part of protein platforms at the cell surface, PrPC may bridge extracellular matrix molecules and/or membrane proteins to intracellular signaling pathways.Key words: prion protein, PrPC, extracellular matrix, cell adhesion molecules, neuritogenesis, p59fyn, Ca2+Initially, the discovery that the prion protein was the major, if not the unique, component of the prion agent causing transmissible spongiform encephalopathies (TSE)1 has placed the protein in an extremely unfavorable light. Thereafter, however, a wealth of evidence has supported the notion that the protein positively influences several aspects of the cell physiology, and that its duality—in harboring both lethal and beneficial potentials—could be rationalized in terms of a structural switch. Indeed, the protein exists in at least two conformational states: the cellular, α helix-rich isoform, PrPC, and the prion-associated β sheet-rich isoform, PrPSc.2 If it is now unquestionable that the presence of PrPC in the cell is mandatory for prion replication and neurotoxicity to occur,3,4 nonetheless its physiologic function is still debatable, despite the long lasting effort, and the numerous, frequently genetically advanced, animal and cell model systems dedicated to the issue. From these studies the picture of an extremely versatile protein has emerged, whereby PrPC acts in the cell defense against oxidative and apoptotic challenges, but also in cell adhesion, proliferation and differentiation, and in synaptic plasticity.5,6 In an effort to converge these multiple propositions in an unifying functional model, different murine lines devoid of PrPC have been studied. These animals, however, displayed no obvious phenotype,79 suggesting that either PrPC is dispensable during development and adult life or that compensative mechanisms mask the loss of PrPC function in these paradigms. Thus, identifying the exact role of PrPC in the cell would not only resolve an important biological question, but would also help elucidate the cellular steps of prion pathogenesis necessary for designing early diagnostic tools and therapeutic strategies for TSE.As is often the case, the employment of a model system unprecedented in prion research has recently disclosed a most interesting scenario with regards to PrPC physiology, having unravelled, for the first time, a lethal phenotype linked to the absence of the protein.10 The paradigm is the zebrafish, which expresses two PrPC isoforms (PrP1 and PrP2). Similarly to mammalian PrPC, they are glycosylated and attached to the external side of the plasma membrane through a glycolipid anchor. PrP1 and PrP2 are, however, expressed in distinct time frames of the zebrafish embryogenesis. Accordingly, the knockdown of the PrP1, or PrP2, gene very early in embryogenesis impaired development at different stages, bypassing putative compensatory mechanisms. By focusing on PrP1, Malaga-Trillo et al. showed that the protein was essential for cell adhesion, and that this event occurred through PrP1 homophilic trans-interactions and signaling. This comprised activation of the Src-related tyrosine (Tyr) kinase p59fyn, and, possibly, Ca2+ metabolism, leading to the regulation of the trafficking of E-cadherin, a member of surface-expressed cell adhesion molecules (CAMs) responsible for cell growth and differentiation.11 It was also reported that overlapping PrP1 functions were performed by PrPCs from other species, while the murine PrPC was capable to replace PrP1 in rescuing, at least in part, the knockdown developmental phenotype. Apart from providing the long-sought proof for a vital role of PrPC, the demonstration that a mammalian isoform corrected the lethal zebrafish phenotype strongly reinforces previous results—mainly obtained in a variety of mammalian primary neurons and cell lines—pointing to a functional interplay of PrPC with CAMs, or extra cellular matrix (ECM) proteins, and cell signaling, to promote neuritogenesis and neuronal survival. A revisit of these data is the main topic of the present minireview.As mentioned, the capacity of PrPC to act as a cell adhesion, or recognition, molecule, and to entertain interactions with proteins implicated in growth and survival, has already been reported for the mammalian PrPC. A case in point is the interaction, both in cis- and trans-configurations, with the neuronal adhesion protein N-CAM12 that led to neurite outgrowth.13 Like cadherins, N-CAM belongs to the CAM superfamily. Following homo- or heterophylic interactions, it can not only mediate adhesion of cells, or link ECM proteins to the cytoskeleton, but also act as a receptor to transduce signals ultimately resulting in modulating neurite outgrowth, neuronal survival and synaptic plasticity.11 Another example is the binding of PrPC to laminin, an ECM heterotrimeric glycoprotein, which induced neuritogenesis together with neurite adhesion and maintenance,14,15 but also learning and memory consolidation.16 Further, it has been described that PrPC interacted with the mature 67 kDa-receptor (67LR) (and its 37 kDa-precursor) for laminin, and with glycosamminoglycans (GAGs), each of which is involved in neuronal differentiation and axon growth.1721 More recently, Hajj et al.22 have reported that the direct interaction of PrPC with another ECM protein, vitronectin, could accomplish the same process, and that the absence of PrPC could be functionally compensated by the overexpression of integrin, another laminin receptor.23 Incidentally, the latter finding may provide a plausible explanation for the absence of clear phenotypes in mammalian PrP-null paradigms. By exposing primary cultured neurons to recombinant PrPs, others have shown that trans-interactions of PrPC are equally important for neuronal outgrowth,24,25 including the formation of synaptic contacts.25 Finally, it has been demonstrated that the binding of PrPC with the secreted co-chaperone stress-inducible protein 1 (STI1) stimulated neuritogenesis.26 This same interaction had also a pro-survival effect, as did the interaction of PrPC with its recombinant form.24 Notably, the involvement of PrPC in cell protection has been heightened by experiments with whole animals. By applying transient or permanent focal cerebral ischemia to the animals, it was found that their reduced brain damage correlated with spontaneous or adenoviral-mediated, upregulation of PrPC,2729 (reviewed in ref. 30), and that PrPC deficiency aggravated their ischemic brain injury.30,31 Thus, now that data are available from phylogenetically distant paradigms (zebrafish and mammalian model systems), it acquires more solid grounds the advocated engagement of PrPC in homo/heterophilic cis/trans interactions to trigger signaling events aiming at neuronal—or, in more general terms, cell—survival and neuritogenesis. The latter notion is consistent with the delayed maturation of different types of PrPC-less neurons, observed both in vitro and in vivo.32,33If one assumes that the interaction of PrPC with multiple partners (45 for PrPC and PrPSc, as reviewed in Aguzzi et al.,5 or 46 considering the homophylic interaction) are all functionally significant, the most immediate interpretation of this “sticky” behavior entails that PrPC acts as a scaffolding protein in different membrane protein complexes.5,6 Each complex could then activate a specific signaling pathway depending on the type and maturation of cells, the expression and glycosylation of PrPC, and availability of extra- and intra-cellular signaling partners. At large, all these signals have been shown to be advantageous to the cell. However, because in a cell only a subtle line divides the “good” from the “bad,” instances can be envisioned in which a pro-life signal turns into a pro-death signal. A typical example of this possibility is glutamate excitotoxicity resulting in dangerous, glutamate receptor-linked, Ca2+ overload. Likewise, an excessive or over-stimulated signal elicited by PrPC, or by the putative complex housing the protein could become noxious to the cell. This possibility may explain why the massive expression of PrPC caused degeneration of the nervous system,34 and of skeletal muscles,34,35 in transgenic animals. More intriguing is the finding that—in a mouse line expressing anchorless PrPC—PrPSc was capable to replicate without threatening the integrity of neurons.36 This may suggest that native membrane-bound PrPC acts as, or takes part into, a “receptor for PrPSc”, and that lasting PrPSc-PrPC interactions distort the otherwise beneficial signal of the protein/complex and cause neurodegeneration.37 Consistent with this hypothesis is the finding that the in vivo antibody-mediated ligation of PrPC provoked apoptosis of the antibody-injected brain area.38 Speculatively, the action of N-terminally, or N-proximally truncated PrPs whose expression in PrP-less transgenic mice induced extensive neurodegeneration,3941 may be traced back to the same hyper-activation of PrPC signaling. Possibly, this may hold true also for the synaptic impairment that, recorded only in PrPC-expressing neurons, was attributed to the binding of amyloid beta (Aβ) peptide oligomers implicated in Alzheimer disease, to PrPC.42,43But which is (are) the cellular signaling pathway(s) conveyed by the engagement of PrPC in different signaling complexes? In line with its multifaceted behavior, several intracellular effectors have been proposed, including p59fyn, mitogen-activated kinases (MAPK) Erk1/2, PI3K/Akt and cAMP-PKA. p59fyn is the most reported downstream effector, suggesting that, in accordance with its behavior, p59fyn could serve as the sorting point for multiple incoming and outgoing signals also in the case of PrPC. The initial evidence of the PrPC-p59fyn connection came from cells subjected to antibody-mediated cross-linking of PrPC.44 Later, it was shown that the PrPC-p59fyn signal converged to Erk1/2 through a pathway dependent on (but also independent of) reactive oxygen species generated by NADPH oxidase.45 A PrPC-dependent activation of p59fyn13,25 and Erk1/2 (but also of PI3K and cAMP-PKA)24 was evident in other neuronal cell paradigms and consistent with the almost ubiquitous expression of PrPC, in non-neuronal cells such as Jurkat and T cells.46 Not to forget that in zebrafish embryonic cells activated p59fyn was found in the same focal adhesion sites harboring PrP1.10 Regarding the activation of the ERK1/2 pathway promoted by the PrPC-STI1 complex, and leading to neuritogenesis, the role of p59fyn was not investigated.26 The same holds true for the transduction of a neuroprotective signal by the PrPC-STI1 complex involving the cAMP-PKA pathway.26 Interestingly, this is not the only example reporting that engagement of PrPC activates simultaneously two independent pathways. In fact, possibly after transactivating the receptor for the epidermal growth factor, the antibody-mediated clustering of PrPC was shown to impinge on both the Erk1/2 pathway, and on a protein (stathmin) involved in controlling microtubule dynamics.47Yet, if p59fyn is implicated in mammalian PrPC-activated signaling cascade, a protein linking extracellular PrPC to p59fyn is needed, given the attachment of the enzyme to the inner leaflet of the plasma membrane through palmitoylated/myristoylated anchors. In this, the PrPC partner N-CAM (isoform 140) seems ideal to fulfill the task, given that p59fyn is part of N-CAM-mediated signaling. Indeed, after recruitment of N-CAM to lipid rafts—which may also depend on PrPC,13—together with the receptor protein Tyr phosphatase α (RPTPα), the Tyr-phosphate removing activity of RPTPα allows the subsequent activation of p59fyn through an autophosphorylation step.48 This event recruits and activates the focal adhesion kinase (FAK),11 another non-receptor Tyr kinase. Finally, formation of the FAK-p59fyn complex triggers neuritogenesis through both Erk1/2 and PI3K/Akt pathways.49,50 Parenthetically, the FAK-p59fyn and PI3K/Akt connection would be suitable to explain why aggravation of ischemic brain injury in PrP-deficient brains was linked to a depressed Akt activation.31 FAK-p59fyn complex, however, may be also involved in the signal triggered by the still mysterious PrPC partner, 67LR. This protein was reported not only to act as a laminin receptor but also to facilitate the interaction of laminin with integrins,51 thereby possibly activating (through integrins) FAK-p59fyn-regulated pathways.49 Conversely, other data have supported the candidature of caveolin-1 for coordinating the signal that from PrPC reaches Erk1/2 through p59fyn.44,45,52 Further scrutiny of this route has shown that it comprised players such as laminin and integrins (upstream), FAK-p59fyn, paxillin and the Src-homology-2 domain containing adaptor protein (downstream), and that caveolin-1, a substrate of the FAK-p59fyn complex, facilitated the interaction of these signaling partners by recruiting them in caveolae-like membrane domains.53For the relevance they bear, we need to acknowledge recent propositions supporting the commitment of PrPC with proteins whose function is unrelated from the above-mentioned cell adhesion or ECM molecules; namely, the β-site amyloid precursor protein (APP) cleaving enzime (BACE1) and the N-methyl-D-aspartate (NMDA)-receptor. BACE1 is a proteolytic enzyme involved in Aβ production. It has been shown that overexpressed PrPC restricted, while depletion of PrPC increased the access of BACE1 to APP, possibly because PrPC interacts with BACE1 via GAGs.54 Thus, native PrPC reduces the production of Aβ peptides. A beneficial effect of PrPC was also highlighted by Khosravani et al.55 showing that, by physically associating with the subunit 2D of the NMDA-receptor, PrPC attenuated neuronal Ca2+ entry and its possible excitotoxic effect. This clear example for the control of PrPC on Ca2+ metabolism is particularly intriguing in light of previous reports linking Ca2+ homeostasis to PrPC pathophysiology (reviewed in ref. 56). Also, it is important to mention that a few partners of PrPC or downstream effectors may initiate signals that increase intracellular Ca2+, and that, in turn, local Ca2+ fluctuations regulate some of the afore-mentioned pathways.11,49,57,58In conclusion, although still somehow speculative, the implication of Ca2+ in PrPC-dependent pathways raises the possibility that the different input signals originating from the interaction of PrPC with diverse partners may all converge to the universal, highly versatile Ca2+ signaling. Were indeed this the case, then clearly the acting of PrPC as Harlequin, the famous character of the 18th century Venetian playwright Carlo Goldoni, who struggles to fill the orders of two masters, would be merely circumstantial.  相似文献   

8.
The tuatara, Sphenodon punctatus, known from 32 small islands around New Zealand, has often been noted as a classic ‘living fossil’ because of its apparently close resemblance to its Mesozoic forebears and because of a long, low‐diversity history. This designation has been disputed because of the wide diversity of Mesozoic forms and because of derived adaptations in living Sphenodon. We provide a testable definition for ‘living fossils’ based on a slow rate of lineage evolution and a morphology close to the centroid of clade morphospace. We show that through their history since the Triassic, rhynchocephalians had heterogeneous rates of morphological evolution and occupied wide morphospaces during the Triassic and Jurassic, and these then declined in the Cretaceous. In particular, we demonstrate that the extant tuatara underwent unusually slow lineage evolution, and is morphologically conservative, being located near the centre of the morphospace for all Rhynchocephalia.  相似文献   

9.
The identification of an association between polymorphisms of the prion protein (PrP) gene and susceptibility to scrapie has enabled the development of breeding programmes to increase natural resistance to scrapie. It is, however, imperative to identify if such selection would affect important reproduction and production traits. The objective of this study was to determine if there is a relationship between polymorphisms at codons 136, 154 and 171 of the PrP gene and ovulation rate or litter size in sheep. Data were collected from a mixed-aged flock of Belclare ewes, over a 9-year period. Ovulation rate was determined annually using laparoscopy by counting the number of corpora lutea at each of two consecutive oestrous cycles, one immediately before and one after mating (2418 records from 366 ewes). Litter size was recorded at parturition (875 records from 353 ewes). The five common PrP alleles were present in the population and 14 PrP genotypes were represented among the animals studied. There was no significant overall effect of PrP genotype on ovulation rate or litter size and pairwise comparisons among genotypes did not reveal any significant differences for either trait. These data suggest that breeding programmes based on selection for specific polymorphisms of the PrP gene will not influence ovulation rate or litter size, at least in the breed studied.  相似文献   

10.
In a recent paper written by Hilbe et al (BMC vet res, 2009), the nature and specificity of the prion protein deposition in the kidney of feline species affected with feline spongiform encephalopathy (FSE) were clearly considered doubtful. This article was brought to our attention because we published several years ago an immunodetection of abnormal prion protein in the kidney of a cheetah affected with FSE. At this time we were convinced of its specificity but without having all the possibilities to demonstrate it. As previously published by another group, the presence of abnormal prion protein in some renal glomeruli in domestic cats affected with FSE is indeed generally considered as doubtful mainly because of low intensity detected in this organ and because control kidneys from safe animals present also a weak prion immunolabelling. Here we come back on these studies and thought it would be helpful to relay our last data to the readers of BMC Vet res for future reference on this subject.  相似文献   

11.
《朊病毒》2013,7(5-6):315-319
ABSTRACT

Although colocalization of amyloid β (Aβ) with prion protein (PrP) in the kuru plaque has previously been observed in the brain of prion diseases patients, the participating Aβ species has not been identified. Here, we present an immunohistochemical assessment of the brain and spinal cord of a 69-year-old Japanese female patient with Gerstmann-Sträussler-Scheinker disease with a P105L mutation on the PRNP gene (GSS-P105L). Immunohistochemical assessment of serial brain sections was performed using anti-PrP and -Aβ antibodies in the hippocampus, frontal and occipital lobes. She died 69 years after a 21-year clinical course. Immunohistochemistorical examination revealed that ~50% of the kuru plaques in the cerebrum were colocalized with Aβ, and Aβ42 was predominantly observed to be colocalized with PrP-plaques. The Aβ deposition patterns were unique, and distinct from diffuse plaques observed in the normal aging brain or Alzheimer’s disease brain. The spinal cord exhibited degeneration in the lateral corticospinal tract, posterior horn, and fasciculus gracilis. We have demonstrated for the first time that Aβ42, rather than Aβ40, is the main Aβ component associated with PrP-plaques, and also the degeneration of the fasciculus gracilis in the spinal cord in GSS-P105L, which could be associated with specific clinical features of GSS-P105L.  相似文献   

12.
The host-encoded prion protein (PrP) is a component of transmissible amyloid deposited in the brains affected by Gerstmann-Str?ussler syndrome (GSS). Recently GSS in two unrelated Caucasian families has been reported to be linked to an amino acid change in PrP codon 102, proline to leucine (Leu102). However, it has not been clear whether the change is commonly found to GSS regardless of ethnic origin. We report here that Leu102 is also found in all the Japanese GSS patients tested. Interestingly, one French GSS patient was found to have another change, alanine to valine in codon 117 (Val117), instead of Leu102. Our results indicate that Leu102 is closely related to GSS irrespective of ethnic origin, but not the sole mutation related to GSS. Val117 may also be related to GSS.  相似文献   

13.
Lipocalin α1-microglobulin (α1M) is a conserved glycoprotein present in plasma and in the interstitial fluids of all tissues. α1M is linked to a heterogeneous yellow–brown chromophore of unknown structure, and interacts with several target proteins, including α1-inhibitor-3, fibronectin, prothrombin and albumin. To date, there is little knowledge about the interaction sites between α1M and its partners. Here, we report the crystal structure of the human α1M. Due to the crystallization occurring in a low ionic strength solution, the unidentified chromophore with heavy electron density is observed at a hydrophobic inner tube of α1M. In addition, two conserved surface regions of α1M are proposed as putative protein–protein interface sites. Further study is needed to unravel the detailed information about the interaction between α1M and its partners.  相似文献   

14.
In this study we investigated the role of Cu(2+), Mn(2+), Zn(2+), and Al(3+) in inducing defective conformational rearrangements of the recombinant human prion protein (hPrP), which trigger aggregation and fibrillogenesis. The research was extended to the fragment of hPrP spanning residues 82-146, which was identified as a major component of the amyloid deposits in the brain of patients affected by Gerstmann-Str?ussler-Scheinker (GSS) disease. Variants of the 82-146 wild-type subunit [PrP-(82-146)(wt)] were also examined, including entirely, [PrP-(82-146)(scr)], and partially scrambled, [PrP-(82-146)(106)(-)(126scr)] and [PrP-(82-146)(127)(-)(146scr)], peptides. Al(3+) strongly stimulated the conversion of native hPrP into the altered conformation, and its potency in inducing aggregation was very high. Despite a lower rate and extent of prion protein conversion into altered isoforms, however, Zn(2+) was more efficient than Al(3+) in promoting organization of hPrP aggregates into well-structured, amyloid-like fibrillar filaments, whereas Mn(2+) delayed and Cu(2+) prevented the process. GSS peptides underwent the fibrillogenesis process much faster than the full-length protein. The intrinsic ability of PrP-(82-146)(wt) to form fibrillar aggregates was exalted in the presence of Zn(2+) and, to a lesser extent, of Al(3+), whereas Cu(2+) and Mn(2+) inhibited the conversion of the peptide into amyloid fibrils. Amino acid substitution in the neurotoxic core (sequence 106-126) of the 82-146 fragment reduced its amyloidogenic potential. In this case, the stimulatory effect of Zn(2+) was lower as compared to the wild-type peptide; on the contrary Al(3+) and Mn(2+) induced a higher propensity to fibrillation, which was ascribed to different binding modalities to GSS peptides. In all cases, alteration of the 127-146 sequence strongly inhibited the fibrillogenesis process, thus suggesting that integrity of the C-terminal region was essential both to confer amyloidogenic properties on GSS peptides and to activate the stimulatory potential of the metal ions.  相似文献   

15.
Animal experiments and observations in human brains have convincingly shown that sexual differentiation not only concerns the genitalia but also the brain. This has been investigated also in the light of a possible explanation of a presumed biological aetiology of transsexuality. The volume of the central subdivision of the bed nucleus of the stria terminalis, a brain area that is essential for sexual behaviour, has been reported to be larger in men than in women. Additionally, the number of somatostatin expressing neurons in this region was shown to be higher in men than in women. As neuronal production of somatostatin is involved the idea is striking whether somatostatin-receptor density in the cortex of cerebral hemispheres might be related to gender identity. We investigated in vivo the density of somatostatin-receptors in selected regions of the human brain in both sexes by means of receptor scintigraphy. Basal ganglia tracer uptake of 111-In-Pentreotide was equally low in both genders at 0,80% +/ 0,26 (related to tracer uptake of the whole brain layer). Temporal cortex accumulated at 2,9% +/ 1,1 in men and at 2,3% +/ 0,76 in women. Frontal brain region had an uptake of 3,0% +/ 1,4 in male and of 2,5% +/ 1,3 in female. This shows a tendency in males for relatively augmented uptake indicating higher somatostatin receptor density in temporal and frontal cerebral cortex.  相似文献   

16.
Background: Elongation and calcification of the stylohyoid ligament complex may be correlated with Eagle's syndrome. The styloid complex pathogenesis is still being debated. Objective: The aim of this study was to investigate the prevalence of stylohyoid ligament complex elongation in panoramic radiographs of 2252 patients in a Brazilian adult, partially edentulous population, of both sexes. Methods: The radiographs, taken in the Semiology Department at the Dental School in Brazil, were randomly selected from January 1997 to December 2000. The stylohyoid ligament complexes were measured from the cranial base up to the osseous tip of each process. Mineralisation of the complex of more than 25 mm in length on the radiograph was considered to be abnormal. The same operator made all the measurements. The lengths of the areas of mineralisation were recorded, and whether the condition was bilateral was noted. Results: This abnormality was present in both sexes. A calcified complex was found in 451 of the 2252 patients. The majority of these calcified complexes (n = 248; 54.9%) were bilateral. Three hundred and fifty‐six (39.5%) of them were longer than 25 mm (and so were abnormal), and the length varied from 26.1 to 65 mm. (average length = 27.8 mm). Forty (4.4%) of the abnormalities were longer than 50 mm, and of these 36 (90%) were in the 40–59 year age group. The mean length of a mineralised stylohyoid ligament complex in patients in the 60–79 year age group was 32.75 mm. Conclusion: The results suggested that an anatomical variant of the stylohyoid ligament complex was more frequently found in the elderly female population, although this abnormality was present in both sexes. There was a greater tendency for the abnormality to be present in patients between 60 and 79 years of age.  相似文献   

17.
The opportunistic human pathogen Staphylococcus epidermidis is the major cause of nosocomial biomaterial infections. S. epidermidis has the ability to attach to indwelling materials coated with extracellular matrix proteins such as fibrinogen, fibronectin, vitronectin, and collagen. To identify the proteins necessary for S. epidermidis attachment to collagen, we screened an expression library using digoxigenin-labeled collagen as well as two monoclonal antibodies generated against the Staphylococcus aureus collagen-adhesin, Cna, as probes. These monoclonal antibodies recognize collagen binding epitopes on the surface of S. aureus and S. epidermidis cells. Using this approach, we identified GehD, the extracellular lipase originally found in S. epidermidis 9, as a collagen-binding protein. Despite the monoclonal antibody cross-reactivity, the GehD amino acid sequence and predicted structure are radically different from those of Cna. The mature GehD circular dichroism spectra differs from that of Cna but strongly resembles that of a mammalian cell-surface collagen binding receptor, known as the alpha(1) integrin I domain, suggesting that they have similar secondary structures. The GehD protein is translated as a preproenzyme, secreted, and post-translationally processed into mature lipase. GehD does not have the conserved LPXTG C-terminal motif present in cell wall-anchored proteins, but it can be detected in lysostaphin cell wall extracts. A recombinant version of mature GehD binds to collagens type I, II, and IV adsorbed onto microtiter plates in a dose-dependent saturable manner. Recombinant, mature GehD protein and anti-GehD antibodies can inhibit the attachment of S. epidermidis to immobilized collagen. These results provide evidence that GehD may be a bi-functional molecule, acting not only as a lipase but also as a cell surface-associated collagen adhesin.  相似文献   

18.
A micromorphological study of archaeological sediments from the early Pleistocene site of 'Ubeidiya (Jordan Valley, Israel) was conducted to provide microenvironmental detail for the hominin occupation contexts and investigate site formation issues. Previous research shows that the hominin groups occupied the marshes and pebbly beaches at the shores of a lake during a regressive period, but given that some portions of the lithic and faunal assemblages are abraded and others fresh, there remains a question of whether the archaeological assemblages are in situ or reworked, and if reworked, by what mechanisms and from where. The rates of sedimentation within the regressive cycle, by which we can learn about the frequency and duration of exposed surfaces amenable for hominin occupation is also unknown. Finally, the artificial nature of some of the pebbly layers has been questioned. The micromorphological analysis yielded the identification of twelve microfacies; the majority of these represent fluvially derived floodplain soils or distal mudflow deposits, and a minor number are sediments of lacustrine origin: mudflats and shallow subaqueous sediments. These represent the natural habitats of the 'Ubeidiya hominins and might serve as a reference to similar contexts of other early hominin sites. The sedimentary model proposed here entails the rapid deposition of fluvially derived low-energy sediments at and around the shoreline, followed by prolonged periods of exposure, during which surfaces stabilized within a relatively wet, marshy environment. This interpretation suggests that the abraded portions of the archaeological assemblages are a result of prolonged surface exposure rather than high-energy transport from a distant source or to wave reworking at the shoreline, and supports the consideration of these assemblages as archaeological palimpsests, with locally reworked fresh and abraded elements. No micromorphological evidence supporting anthropogenic agency in the formation of the pebbly layers was found. The entire regressive cycle entailed unvarying climatic conditions with seasonal fluctuations and episodic lacustrine incursions, and with a trend towards arid conditions in the end.  相似文献   

19.
20.
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