Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
1.
Bioengineering (Basel) ; 11(3)2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38534516

RESUMO

The cellular prion protein (PrPc) is a cell surface glycoprotein that is highly expressed in a variety of cancer tissues in addition to the nervous system, and its elevated expression is correlated to poor prognosis in many cancer patients. Our team previously found that patients with colorectal cancer (CRC) with high-level PrPc expression had significantly poorer survival than those with no or low-level PrPc expression. Mouse antibodies for PrPc inhibited tumor initiation and liver metastasis of PrPc-positive human CRC cells in mouse model experiments. PrPc is a candidate target for CRC therapy. In this study, we newly cloned a mouse anti-PrPc antibody (Clone 6) and humanized it, then affinity-matured this antibody using a CHO cell display with a peptide antigen and full-length PrPc, respectively. We obtained two humanized antibody clones with affinities toward a full-length PrPc of about 10- and 100-fold of that of the original antibody. The two humanized antibodies bound to the PrPc displayed significantly better on the cell surface than Clone 6. Used for Western blotting and immunohistochemistry, the humanized antibody with the highest affinity is superior to the two most frequently used commercial antibodies (8H4 and 3F4). The two new antibodies have the potential to be developed as useful reagents for PrPc detection and even therapeutic antibodies targeting PrPc-positive cancers.

2.
J Cancer ; 15(6): 1593-1602, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38370370

RESUMO

Background: Oral squamous cell carcinoma (OSCC) is frequently diagnosed at an advanced stage, and the high mortality of patients is mainly due to the delay of diagnosis. Cellular prion protein (PrPC) contributes to the occurrence and development of many malignant tumors. However, little has been known about the clinical and diagnostic value of PrPC in OSCC. This study investigated the levels of PrPC in the saliva and serum of patients with OSCC, OPMD and control group and their diagnostic value. Methods: The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and Clinical Proteome Tumor Analysis Consortium (CPTAC) databases were analyzed to evaluate the expression of human prion protein gene (PRNP) mRNA and PrPC in OSCC. Enzyme-linked Immunosorbent Assay (ELISA) was utilized to detect the expression of PrPC in saliva and serum samples of OSCC, OPMD and control groups. Furthermore, diagnostic value and clinical significance of PrPC in OSCC was identified. Protein-protein interaction (PPI) network was constructed by STRING. GO and KEGG analysis were performed by ClusterProfiler. Results: The levels of PRNP mRNA and PrPC in OSCC were significantly higher than those in the control group from databases (P<0.05). Besides, salivary and serum PrPC of OSCC patients showed increased levels compared with OPMD and control groups (P<0.05). The expression of salivary and serum PrPC of OSCC was correlated with the degree of differentiation (P<0.05), and the expression of PrPC from CPTAC was related to tumor stage of OSCC (P<0.05). The areas under the diagnostic curves (AUCs) of salivary and serum PrPC were 0.807 and 0.671, respectively. GO and KEGG analysis revealed that PrPC might be related to cell adhesion, cell differentiation, signal transduction and apoptosis, and participate in the pathways of focal adhesion, PI3K-Akt signaling pathway and ECM- receptor interaction in OSCC. Conclusion: PrPC in saliva and serum may be a potential biomarker for early diagnosis of OSCC.

3.
JMIR Form Res ; 8: e45573, 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38214964

RESUMO

BACKGROUND: Twitter is a common platform for people to share opinions, discuss health-related topics, and engage in conversations with a wide audience. Twitter users frequently share health information related to chronic diseases, mental health, and general wellness topics. However, sharing health information on Twitter raises privacy concerns as it involves sharing personal and sensitive data on a web-based platform. OBJECTIVE: This study aims to adopt an interactive approach and develop a model consisting of privacy concerns related to web-based vendors and web-based peers. The research model integrates the 4 dimensions of concern for information privacy that express concerns related to the practices of companies and the 4 dimensions of peer privacy concern that reflect concerns related to web-based interactions with peers. This study examined how this interaction may affect individuals' information-sharing behavior on Twitter. METHODS: Data were collected from 329 Twitter users in the United States using a web-based survey. RESULTS: Results suggest that privacy concerns related to company practices might not significantly influence the sharing of general health information, such as details about hospitals and medications. However, privacy concerns related to companies and third parties can negatively shape the disclosure of specific health information, such as personal medical issues (ß=-.43; P<.001). Findings show that peer-related privacy concerns significantly predict sharing patterns associated with general (ß=-.38; P<.001) and specific health information (ß=-.72; P<.001). In addition, results suggest that people may disclose more general health information than specific health information owing to peer-related privacy concerns (t165=4.72; P<.001). The model explains 41% of the variance in general health information disclosure and 67% in specific health information sharing on Twitter. CONCLUSIONS: The results can contribute to privacy research and propose some practical implications. The findings provide insights for developers, policy makers, and health communication professionals about mitigating privacy concerns in web-based health information sharing. It particularly underlines the importance of addressing peer-related privacy concerns. The study underscores the need to build a secure and trustworthy web-based environment, emphasizing the significance of peer interactions and highlighting the need for improved regulations, clear data handling policies, and users' control over their own data.

4.
Cancers (Basel) ; 15(20)2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37894349

RESUMO

The cellular prion protein (PrPC) is a glycoprotein anchored to the cell surface by glycosylphosphatidylinositol (GPI). PrPC is expressed both in the brain and in peripheral tissues. Investigations on PrPC's functions revealed its direct involvement in neurodegenerative and prion diseases, as well as in various physiological processes such as anti-oxidative functions, copper homeostasis, trans-membrane signaling, and cell adhesion. Recent findings have revealed the ectopic expression of PrPC in various cancers including gastric, melanoma, breast, colorectal, pancreatic, as well as rare cancers, where PrPC promotes cellular migration and invasion, tumor growth, and metastasis. Through its downstream signaling, PrPC has also been reported to be involved in resistance to chemotherapy and tumor cell apoptosis. This review summarizes the variance of expression of PrPC in different types of cancers and discusses its roles in their development and progression, as well as its use as a potential target to treat such cancers.

5.
Front Microbiol ; 14: 1175679, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37637115

RESUMO

Introduction: Infection with human T cell lymphotropic virus type 1 (HTLV-1) is endemic in Brazil and is linked with pro-inflammatory conditions including HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), a chronic neuroinflammatory incapacitating disease that culminates in loss of motor functions. The mechanisms underlying the onset and progression of HAM/TSP are incompletely understood. Previous studies have demonstrated that inflammation and infectious agents can affect the expression of cellular prion protein (PrPC) in immune cells. Methods: Here, we investigated whether HTLV-1 infection affected PrPC content in cell lines and primary CD4+cells in vitro using flow cytometry and western blot assays. Results: We found that HTLV-1 infection decreased the expression levels of PrPC and HTLV-1 Orf I encoded p12, an endoplasmic reticulum resident protein also known to affect post-transcriptionally cellular proteins such as MHC-class I and the IL-2 receptor. In addition, we observed a reduced percentage of CD4+ T cells from infected individuals expressing PrPC, which was reflected by IFN type II but not IL-17 expression. Discussion: These results suggested that PrPC downregulation, linked to both HTLV-1 p12 and IFN-γ expression in CD4+ cells, may play a role in the neuropathogenesis of HTLV-1 infection.

6.
J Biol Chem ; 299(8): 105054, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37454740

RESUMO

Neurodegenerative diseases are often characterized by the codeposition of different amyloidogenic proteins, normally defining distinct proteinopathies. An example is represented by prion diseases, where the classical deposition of the aberrant conformational isoform of the prion protein (PrPSc) can be associated with tau insoluble species, which are usually involved in another class of diseases called tauopathies. How this copresence of amyloidogenic proteins can influence the progression of prion diseases is still a matter of debate. Recently, the cellular form of the prion protein, PrPC, has been investigated as a possible receptor of amyloidogenic proteins, since its binding activity with Aß, tau, and α-synuclein has been reported, and it has been linked to several neurotoxic behaviors exerted by these proteins. We have previously shown that the treatment of chronically prion-infected cells with tau K18 fibrils reduced PrPSc levels. In this work, we further explored this mechanism by using another tau construct that includes the sequence that forms the core of Alzheimer's disease tau filaments in vivo to obtain a distinct fibril type. Despite a difference of six amino acids, these two constructs form fibrils characterized by distinct biochemical and biological features. However, their effects on PrPSc reduction were comparable and probably based on the binding to PrPC at the plasma membrane, inhibiting the pathological conversion event. Our results suggest PrPC as receptor for different types of tau fibrils and point out a role of tau amyloid fibrils in preventing the pathological PrPC to PrPSc conformational change.


Assuntos
Doenças Neurodegenerativas , Doenças Priônicas , Príons , Proteínas tau , Humanos , Proteínas Amiloidogênicas , Doenças Priônicas/metabolismo , Proteínas Priônicas , Príons/metabolismo , Proteínas tau/metabolismo
7.
Int J Mol Sci ; 24(7)2023 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-37047224

RESUMO

There is a large unmet medical need to develop disease-modifying treatment options for individuals with age-related degenerative diseases of the central nervous system. The sigma-2 receptor (S2R), encoded by TMEM97, is expressed in brain and retinal cells, and regulates cell functions via its co-receptor progesterone receptor membrane component 1 (PGRMC1), and through other protein-protein interactions. Studies describing functions of S2R involve the manipulation of expression or pharmacological modulation using exogenous small-molecule ligands. These studies demonstrate that S2R modulates key pathways involved in age-related diseases including autophagy, trafficking, oxidative stress, and amyloid-ß and α-synuclein toxicity. Furthermore, S2R modulation can ameliorate functional deficits in cell-based and animal models of disease. This review summarizes the current evidence-based understanding of S2R biology and function, and its potential as a therapeutic target for age-related degenerative diseases of the central nervous system, including Alzheimer's disease, α-synucleinopathies, and dry age-related macular degeneration.


Assuntos
Doença de Alzheimer , Doença por Corpos de Lewy , Receptores sigma , Animais , Doença de Alzheimer/tratamento farmacológico , Receptores sigma/metabolismo , alfa-Sinucleína/metabolismo , Peptídeos beta-Amiloides , Biologia
8.
Food Res Int ; 167: 112636, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37087231

RESUMO

In this study, we compared the heat tolerance parameter (D65℃) values of Salmonella enterica serovar Enteritidis PT 30 (S. Enteritidis ) heat adapted at different degrees (at 42 ℃ for 20-180 min) and cultivated using two methods. The treated group with the highest D65℃ value (LP-42 ℃-60 min) and the untreated groups (Control-TSB and Control-TSA) were subjected to transcriptome analysis. Heat-adaptation increased the D65℃ values of S. Enteritidis by 24.5-60.8%. The D65℃ values of the LP-42 ℃-60 min group (1.85 ± 0.13 min, 7.7% higher) was comparable to that of the Control-TSA. A total of 483 up- and 443 downregulated genes of S. enteritidis were identified in the LP-42 ℃-60 min group (log2fold change > 1, adjusted p-value < 0.05). Among these genes, 5 co-expressed and 15 differentially expressed genes in the LP-42 ℃-60 min and Control-TSA grops possibly contributed to the high D65℃ values of S. Enteritidis . The Rpo regulon was involved in the heat adaptation of S. Enteritidis , as evidenced by the significant upregulation of rpoS, rpoN, and rpoE. KEGG enrichment pathways, such as biosynthesis of secondary metabolites, tricarboxylic acid, and ribosomes were identified and mapped to reveal the molecular mechanisms of S. enteritidis during heat adaptation. This study quantified the enhanced heat tolerance of S. Enteritidis heat adapted at different degrees of heat-adaptation. The results of this study may serve as a basis for elucidating the molecular mechanisms underlying the enhanced heat tolerance at the transcriptome level.


Assuntos
Salmonella enterica , Termotolerância , Salmonella enteritidis/genética , Salmonella enterica/genética , Termotolerância/genética , Temperatura Alta , Perfilação da Expressão Gênica
10.
Int J Mol Sci ; 24(2)2023 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-36674920

RESUMO

The carcinogenesis of glial tumors appears complex because of the many genetic and epigenetic phenomena involved. Among these, cellular prion protein (PrPC) is considered a key factor in cell-death resistance and important aspect implicated in tumorigenesis. Autophagy also plays an important role in cell death in various pathological conditions. These two cellular phenomena are related and share the same activation by specific alterations in the cellular microenvironment. Furthermore, there is an interdependence between autophagy and prion activity in glioma tumorigenesis. Glioma is one of the most aggressive known cancers, and the fact that such poorly studied processes as autophagy and PrPC activity are so strongly involved in its carcinogenesis suggests that by better understanding their interaction, more can be understood about its origin and treatment. Few studies in the literature relate these two cellular phenomena, much less try to explain their combined activity and role in glioma carcinogenesis. In this study, we explored the recent findings on the molecular mechanism and regulation pathways of autophagy, examining the role of PrPC in autophagy processes and how they may play a central role in glioma tumorigenesis. Among the many molecular interactions that PrP physiologically performs, it appears that processes shared with autophagy activity are those most implicated in glial tumor carcinogeneses such as activity on MAP kinases, PI3K, and mTOR. This work can be supportive and valuable as a basis for further future studies on this topic.


Assuntos
Glioma , Proteínas PrPC , Príons , Humanos , Proteínas Priônicas , Príons/metabolismo , Glioma/genética , Autofagia , Carcinogênese/genética , Transformação Celular Neoplásica/genética , Proteínas PrPC/metabolismo , Microambiente Tumoral
11.
Cell Tissue Res ; 392(1): 269-283, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35107622

RESUMO

The properties of infectious prions and the pathology of the diseases they cause are dependent upon the unique conformation of each prion strain. How the pathology of prion disease correlates with different strains and genetic backgrounds has been investigated via in vivo assays, but how interactions between specific prion strains and cell types contribute to the pathology of prion disease has been dissected more effectively using in vitro cell lines. Observations made through in vivo and in vitro assays have informed each other with regard to not only how genetic variation influences prion properties, but also how infectious prions are taken up by cells, modified by cellular processes and propagated, and the cellular components they rely on for persistent infection. These studies suggest that persistent cellular infection results from a balance between prion propagation and degradation. This balance may be shifted depending upon how different cell lines process infectious prions, potentially altering prion stability, and how fast they can be transported to the lysosome. Thus, in vitro studies have given us a deeper understanding of the interactions between different prions and cell types and how they may influence prion disease phenotypes in vivo.


Assuntos
Doenças Priônicas , Príons , Humanos , Príons/metabolismo , Doenças Priônicas/genética , Doenças Priônicas/metabolismo , Linhagem Celular
12.
J Biol Chem ; 299(2): 102823, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36565989

RESUMO

The prion protein (PrPC) is subjected to several conserved endoproteolytic events producing bioactive fragments that are of increasing interest for their physiological functions and their implication in the pathogenesis of prion diseases and other neurodegenerative diseases. However, systematic and comprehensive investigations on the full spectrum of PrPC proteoforms have been hampered by the lack of methods able to identify all PrPC-derived proteoforms. Building on previous knowledge of PrPC endoproteolytic processing, we thus developed an optimized Western blot assay able to obtain the maximum information about PrPC constitutive processing and the relative abundance of PrPC proteoforms in a complex biological sample. This approach led to the concurrent identification of the whole spectrum of known endoproteolytic-derived PrPC proteoforms in brain homogenates, including C-terminal, N-terminal and, most importantly, shed PrPC-derived fragments. Endoproteolytic processing of PrPC was remarkably similar in the brain of widely used wild type and transgenic rodent models, with α-cleavage-derived C1 representing the most abundant proteoform and ADAM10-mediated shedding being an unexpectedly prominent proteolytic event. Interestingly, the relative amount of shed PrPC was higher in WT mice than in most other models. Our results indicate that constitutive endoproteolytic processing of PrPC is not affected by PrPC overexpression or host factors other than PrPC but can be impacted by PrPC primary structure. Finally, this method represents a crucial step in gaining insight into pathophysiological roles, biomarker suitability, and therapeutic potential of shed PrPC and for a comprehensive appraisal of PrPC proteoforms in therapies, drug screening, or in the progression of neurodegenerative diseases.


Assuntos
Western Blotting , Fragmentos de Peptídeos , Proteínas PrPC , Proteólise , Animais , Camundongos , Western Blotting/métodos , Doenças Priônicas/metabolismo , Doenças Priônicas/patologia , Doenças Priônicas/fisiopatologia , Proteínas PrPC/química , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Encéfalo/metabolismo
13.
J Mol Med (Berl) ; 100(10): 1405-1425, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36056255

RESUMO

Cellular prion protein (PrPC) is a highly conserved glycoprotein, present both anchored in the cell membrane and soluble in the extracellular medium. It has a diversity of ligands and is variably expressed in numerous tissues and cell subtypes, most notably in the central nervous system (CNS). Its importance has been brought to light over the years both under physiological conditions, such as embryogenesis and immune system homeostasis, and in pathologies, such as cancer and neurodegenerative diseases. During development, PrPC plays an important role in CNS, participating in axonal growth and guidance and differentiation of glial cells, but also in other organs such as the heart, lung, and digestive system. In diseases, PrPC has been related to several types of tumors, modulating cancer stem cells, enhancing malignant properties, and inducing drug resistance. Also, in non-neoplastic diseases, such as Alzheimer's and Parkinson's diseases, PrPC seems to alter the dynamics of neurotoxic aggregate formation and, consequently, the progression of the disease. In this review, we explore in detail the multiple functions of this protein, which proved to be relevant for understanding the dynamics of organism homeostasis, as well as a promising target in the treatment of both neoplastic and degenerative diseases.


Assuntos
Neoplasias , Doenças Neurodegenerativas , Proteínas PrPC , Sistema Nervoso Central/metabolismo , Humanos , Células-Tronco Neoplásicas/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo
14.
Part Fibre Toxicol ; 19(1): 48, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35840975

RESUMO

BACKGROUND: Epidemiological emerging evidence shows that human exposure to some nanosized materials present in the environment would contribute to the onset and/or progression of Alzheimer's disease (AD). The cellular and molecular mechanisms whereby nanoparticles would exert some adverse effects towards neurons and take part in AD pathology are nevertheless unknown. RESULTS: Here, we provide the prime evidence that titanium dioxide (TiO2) and carbon black (CB) nanoparticles (NPs) bind the cellular form of the prion protein (PrPC), a plasma membrane protein well known for its implication in prion diseases and prion-like diseases, such as AD. The interaction between TiO2- or CB-NPs and PrPC at the surface of neuronal cells grown in culture corrupts PrPC signaling function. This triggers PrPC-dependent activation of NADPH oxidase and subsequent production of reactive oxygen species (ROS) that alters redox equilibrium. Through PrPC interaction, NPs also promote the activation of 3-phosphoinositide-dependent kinase 1 (PDK1), which in turn provokes the internalization of the neuroprotective TACE α-secretase. This diverts TACE cleavage activity away from (i) TNFα receptors (TNFR), whose accumulation at the plasma membrane augments the vulnerability of NP-exposed neuronal cells to TNFα -associated inflammation, and (ii) the amyloid precursor protein APP, leading to overproduction of neurotoxic amyloid Aß40/42 peptides. The silencing of PrPC or the pharmacological inhibition of PDK1 protects neuronal cells from TiO2- and CB-NPs effects regarding ROS production, TNFα hypersensitivity, and Aß rise. Finally, we show that dysregulation of the PrPC-PDK1-TACE pathway likely occurs in the brain of mice injected with TiO2-NPs by the intra-cerebro-ventricular route as we monitor a rise of TNFR at the cell surface of several groups of neurons located in distinct brain areas. CONCLUSION: Our in vitro and in vivo study thus posits for the first time normal cellular prion protein PrPC as being a neuronal receptor of TiO2- and CB-NPs and identifies PrPC-coupled signaling pathways by which those nanoparticles alter redox equilibrium, augment the intrinsic sensitivity of neurons to neuroinflammation, and provoke a rise of Aß peptides. By identifying signaling cascades dysregulated by TiO2- and CB-NPs in neurons, our data shed light on how human exposure to some NPs might be related to AD.


Assuntos
Doença de Alzheimer , Nanopartículas , Príons , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/patologia , Animais , Homeostase , Humanos , Camundongos , Nanopartículas/toxicidade , Neurônios/patologia , Proteínas Priônicas/metabolismo , Príons/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fuligem/toxicidade , Titânio , Fator de Necrose Tumoral alfa/metabolismo
15.
Cancer Lett ; 540: 215725, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35561877

RESUMO

Cancer stem cells (CSCs) are a subpopulation of cancer cells that drive tumour progression and metastasis. Anti-CSC strategies represent new targets for cancer therapies. However, CSCs are highly plastic and heterogeneous, making validation and targeting difficult without bona fide markers that define their identity, especially in a clinical setting. Here, we report that a leucine-rich repeat containing G protein-coupled receptor 4 (LGR4) cooperates with CD44 and PrPc; the latter contributes significantly to metastatic capacity and defines the stemness characteristics of colorectal CSCs. CD44+PrPc+LGR4+ cells effectively developed into organoids and, when transplanted, generated orthotopic and metastatic tumours. Importantly, targeting LGR4 and PrPc with lentiviral shRNAs inhibited organoid development and the growth of orthotopic tumours by inhibiting Wnt/ß-catenin signalling. Thus, our study offers a novel therapeutic strategy that simultaneously targets CSC stemness and metastatic properties.


Assuntos
Neoplasias Colorretais , Neoplasias Hepáticas , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/patologia , Receptores Acoplados a Proteínas G/genética , Via de Sinalização Wnt
16.
J Alzheimers Dis ; 87(3): 1189-1203, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35431249

RESUMO

BACKGROUND: Amyloid-ß (Aß) oligomers induce the overproduction of phosphorylated tau and neurodegeneration. These cascades gradually cause cognitive impairment in Alzheimer's disease (AD). While each pathological event in AD has been studied in detail separately, the spatial and temporal relationships between pathological events in AD remain unclear. OBJECTIVE: We demonstrated that lipid rafts function as a common platform for the pathological cascades of AD. METHODS: Cellular and synaptosomal lipid rafts were prepared from the brains of Aß amyloid model mice (Tg2576 mice) and double transgenic mice (Tg2576 x TgTauP301L mice) and longitudinally analyzed. RESULTS: Aß dimers, the cellular prion protein (PrPc), and Aß dimer/PrPc complexes were detected in the lipid rafts. The levels of Fyn, the phosphorylated NR2B subunit of the N-methyl-D-aspartate receptor, glycogen synthase kinase 3ß, total tau, phosphorylated tau, and tau oligomers increased with Aß dimer accumulation in both the cellular and synaptosomal lipid rafts. Increases in the levels of these molecules were first seen at 6 months of age and corresponded with the early stages of Aß accumulation in the amyloid model mice. CONCLUSION: Lipid rafts act as a common platform for the progression of AD pathology. The findings of this study suggest a novel therapeutic approach to AD, involving the modification of lipid raft components and the inhibition of their roles in the sequential pathological events of AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Microdomínios da Membrana , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/patologia , Camundongos , Camundongos Transgênicos , Fosforilação , Proteínas Priônicas/análise , Proteínas Priônicas/metabolismo , Proteínas tau/metabolismo
17.
Int J Biol Sci ; 18(4): 1755-1772, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35280685

RESUMO

Endometriosis (EMs) is characterized as an estrogen-dependent disease. Whereas, the underlying mechanism for activated estrogen biosynthesis in EMs lesions is largely unknown. We analyzed cholesterol metabolism and estrogen biosynthesis condition of EMs lesions by biological information analysis of GEO datasets, and further verified both in vitro and in vivo by constructing EMs models with uterus fragments from donors of PRNP knockout mouse (Prnp-/-, KO119), Octapeptide repeat region of PRNP knockout mouse (KO120) and PRNP transgenic mouse (Tg20). We found that transcriptome of cholesterol metabolism and estrogen-converting enzymes were disturbed in EMs patients, and cellular cholesterol concentration and local estradiol level were substantially increased in EMs lesions, as well as the high level of prion (PrPC, encoded by PRNP). Notably, 17-ß estradiol stimulation significantly up-regulated PrPC expression in endometrial stromal cells (ESC) and PrPC promoted the proliferative, migratory and invasive abilities of ESC, and was further verified to accelerate EMs progression in mouse models. More importantly, PrPC promoted cholesterol accumulation and activated estrogen biosynthesis of ESC in a PPARα pathway-dependent manner. Taken together, this study suggests that PrPC-cholesterol metabolism/estrogen biosynthesis contributes to the progression of EMs by negatively regulating PPARα pathway, and could be potential therapeutic targets for EMs intervention.


Assuntos
Endometriose , Animais , Endometriose/genética , Endometriose/metabolismo , Estradiol , Estrogênios/metabolismo , Feminino , Humanos , Camundongos , PPAR alfa/metabolismo , Células Estromais/metabolismo
18.
Biochim Biophys Acta Mol Cell Res ; 1869(6): 119240, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35192891

RESUMO

The prion protein is a multifunctional protein that exists in at least two different folding states. It is subject to diverse proteolytic processing steps that lead to prion protein fragments some of which are membrane-bound whereas others are soluble. A multitude of ligands bind to the prion protein and besides proteinaceous binding partners, interaction with metal ions and nucleic acids occurs. Although of great importance, information on structural and functional consequences of prion protein binding to its partners is limited. Here, we will reflect on the structure-function relationship of the prion protein and its binding partners considering the different folding states and prion protein fragments.


Assuntos
Doenças Priônicas , Príons , Humanos , Ligantes , Proteínas Priônicas/genética , Relação Estrutura-Atividade
19.
Biomedicines ; 10(1)2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-35052785

RESUMO

Upon its discovery, Alzheimer's, the neurodegenerative disease that affects many millions of patients in the world, remained without an effective therapy. The first drugs, made available near the end of last century, induced some effects, which remained only marginal. More promising effects are now present, induced by two approaches. Blockers of the enzyme BACE-1 induce, in neurons and glial cells, decreased levels of Aß, the key peptide of the Alzheimer's disease. If administered at early AD steps, the BACE-1 blockers preclude further development of the disease. However, they have no effect on established, irreversible lesions. The extracellular vesicles secreted by mesenchymal stem cells induce therapy effects analogous, but more convenient, than the effects of their original cells. After their specific fusion to target cells, the action of these vesicles depends on their ensuing release of cargo molecules, such as proteins and many miRNAs, active primarily on the cell cytoplasm. Operationally, these vesicles exhibit numerous advantages: they exclude, by their accurate selection, the heterogeneity of the original cells; exhibit molecular specificity due to their engineering and drug accumulation; and induce effective actions, mediated by variable concentrations of factors and molecules and by activation of signaling cascades. Their strength is reinforced by their combination with various factors and processes. The recent molecular and operations changes, induced especially by the stem cell target cells, result in encouraging and important improvement of the disease. Their further development is expected in the near future.

20.
BBA Adv ; 2: 100035, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-37082595

RESUMO

Cellular protein homeostasis (proteostasis) requires an accurate balance between protein biosynthesis, folding, and degradation, and its instability is causally related to human diseases and cancers. Here, we created numerous engineered cancer cell lines targeting APP (amyloid ß precursor protein) and/or PRNP (cellular prion) genes and we showed that APP knocking-down impaired PRNP mRNA level and vice versa, suggesting a link between their gene regulation. PRNPKD, APPKD and PRNPKD/APPKD HeLa cells encountered major difficulties to grow in a 3D tissue-like environment. Unexpectedly, we found a cytoplasmic accumulation of the PrPc protein without PRNP gene up regulation, in both APPKD and APPKO HeLa cells. Interestingly, APP and/or PRNP gene ablation enhanced the chaperone/serine protease HTRA2 gene expression, which is a protein processing quality factor involved in Alzheimer's disease. Importantly, HTRA2 gene silencing decreased PRNP mRNA level and lowered PrPc protein amounts, and conversely, HTRA2 overexpression increased PRNP gene regulation and enhanced membrane-anchored and cytoplasmic PrPc fractions. PrPc, APP and HTRA2 destabilized membrane-associated CD24 protein, suggesting changes in the lipid raft structure. Our data show for the first time that APP and the dual chaperone/serine protease HTRA2 protein could modulate PrPc proteostasis hampering cancer cell behavior.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA