Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 519
Filtrar
1.
Biomolecules ; 13(10)2023 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-37892174

RESUMO

The insulin-degrading enzyme (IDE) is a Zn2+ peptidase originally discovered as the main enzyme involved in the degradation of insulin and other amyloidogenic peptides, such as the ß-amyloid (Aß) peptide. Therefore, a role for the IDE in the cure of diabetes and Alzheimer's disease (AD) has been long envisaged. Anyway, its role in degrading amyloidogenic proteins remains not clearly defined and, more recently, novel non-proteolytic functions of the IDE have been proposed. From a structural point of view, the IDE presents an atypical clamshell structure, underscoring unique enigmatic enzymological properties. A better understanding of the structure-function relationship may contribute to solving some existing paradoxes of IDE biology and, in light of its multifunctional activity, might lead to novel therapeutic approaches.


Assuntos
Doença de Alzheimer , Insulisina , Humanos , Insulisina/química , Insulisina/metabolismo , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/metabolismo , Proteínas Amiloidogênicas , Desenho de Fármacos
2.
J Neuroinflammation ; 20(1): 233, 2023 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-37817156

RESUMO

The insulin-degrading enzyme (IDE) is an evolutionarily conserved zinc-dependent metallopeptidase highly expressed in the brain, where its specific functions remain poorly understood. Besides insulin, IDE is able to cleave many substrates in vitro, including amyloid beta peptides, making this enzyme a candidate pathophysiological link between Alzheimer's disease (AD) and type 2 diabetes (T2D). These antecedents led us to address the impact of IDE absence in hippocampus and olfactory bulb. A specific induction of microgliosis was found in the hippocampus of IDE knockout (IDE-KO) mice, without any effects in neither hippocampal volume nor astrogliosis. Performance on hippocampal-dependent memory tests is influenced by IDE gene dose in 12-month-old mice. Furthermore, a comprehensive characterization of the impact of IDE haploinsufficiency and total deletion in metabolic, behavioral, and molecular parameters in the olfactory bulb, a site of high insulin receptor levels, reveals an unambiguous barcode for IDE-KO mice at that age. Using wildtype and IDE-KO primary microglial cultures, we performed a functional analysis at the cellular level. IDE absence alters microglial responses to environmental signals, resulting in impaired modulation of phenotypic states, with only transitory effects on amyloid-ß management. Collectively, our results reveal previously unknown physiological functions for IDE in microglia that, due to cell-compartment topological reasons, cannot be explained by its enzymatic activity, but instead modulate their multidimensional response to various damaging conditions relevant to aging and AD conditions.


Assuntos
Doença de Alzheimer , Diabetes Mellitus Tipo 2 , Insulisina , Camundongos , Animais , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Insulisina/genética , Insulisina/metabolismo , Insulisina/farmacologia , Microglia/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Encéfalo/metabolismo , Fenótipo
3.
ACS Chem Neurosci ; 14(16): 2935-2943, 2023 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-37498802

RESUMO

The insulin-degrading enzyme (IDE) is an evolutionarily conserved protease implicated in the degradation of insulin and amyloidogenic peptides. Most of the biochemical and biophysical characterization of IDE's catalytic activity has been conducted using solutions containing a single substrate, i.e., insulin or Aß(1-40). IDE's activity toward a particular substrate, however, is likely to be influenced by the presence of other substrates. Here, we show by a kinetic assay based on insulin's helical circular dichroic signal and MALDI TOF mass spectrometry that Aß peptides modulate IDE's activity toward insulin in opposing ways. Aß(1-40) enhances IDE-dependent degradation of insulin, whereas Aß(pyroE3-42), the most pathogenic pyroglutamate-modified Aß peptide in AD, inhibits IDE's activity. Intriguingly, Aß(pyroE3-42) also inhibits IDE's ability to degrade Aß(1-40). Together, our results implicate Aß peptides in the abnormal catabolism of IDE's key substrates.


Assuntos
Insulisina , Insulisina/metabolismo , Peptídeos beta-Amiloides/metabolismo , Insulina/metabolismo
4.
J Clin Lab Anal ; 37(13-14): e24949, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37515308

RESUMO

BACKGROUND: Insulin-degrading enzyme (IDE) is an important gene in studies of the pathophysiology of type 2 diabetes mellitus (T2DM). Recent studies have suggested a possible link between type 2 diabetes mellitus (T2DM) and the pathophysiology of schizophrenia (SZ). At the same time, significant changes in insulin-degrading enzyme (IDE) gene expression have been found in the brains of people with schizophrenia. These findings highlight the need to further investigate the role of IDE in schizophrenia pathogenesis. METHODS: We enrolled 733 participants from the Czech Republic, including 383 patients with schizophrenia and 350 healthy controls. Our study focused on the single nucleotide polymorphism (SNP) rs2421943 in the IDE gene, which has previously been associated with the pathogenesis of Alzheimer's disease. The SNP was analyzed using the PCR-RFLP method. RESULTS: The G allele of the rs2421943 polymorphism was found to significantly increase the risk of developing SZ (p < 0.01) when a gender-based analysis showed that both AG and GG genotypes were associated with a more than 1.55 times increased risk of SZ in females (p < 0.03) but not in males. Besides, we identified a potential binding site at the G allele locus for has-miR-7110-5p, providing a potential mechanism for the observed association. CONCLUSION: Our results confirm the role of the IDE gene in schizophrenia pathogenesis and suggest that future research should investigate the relationship between miRNA and estrogen influence on IDE expression in schizophrenia pathogenesis.


Assuntos
Doença de Alzheimer , Diabetes Mellitus Tipo 2 , Insulisina , Esquizofrenia , Masculino , Feminino , Humanos , Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Mellitus Tipo 2/genética , Esquizofrenia/genética , Insulisina/genética , Insulisina/metabolismo , Genótipo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Polimorfismo de Nucleotídeo Único/genética
5.
Diabetes Obes Metab ; 25(11): 3268-3278, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37493025

RESUMO

AIM: To investigate the use of synthetic preimplantation factor (sPIF) as a potential therapeutic tool for improving glucose-stimulated insulin secretion (GSIS), glucose tolerance and insulin sensitivity in the setting of diabetes. MATERIALS AND METHODS: We used a preclinical murine model of type 2 diabetes (T2D) induced by high-fat diet (HFD) feeding for 12 weeks. Saline or sPIF (1 mg/kg/day) was administered to mice by subcutaneously implanted osmotic mini-pumps for 25 days. Glucose tolerance, circulating insulin and C-peptide levels, and GSIS were assessed. In addition, ß-cells (Min-6) were used to test the effects of sPIF on GSIS and insulin-degrading enzyme (IDE) activity in vitro. The effect of sPIF on GSIS was also tested in human islets. RESULTS: GSIS was enhanced 2-fold by sPIF in human islets ex vivo. Furthermore, continuous administration of sPIF to HFD mice increased circulating levels of insulin and improved glucose tolerance, independently of hepatic insulin clearance. Of note, islets isolated from mice treated with sPIF exhibited restored ß-cell function. Finally, genetic (shRNA-IDE) or pharmacological (6bK) inactivation of IDE in Min-6 abolished sPIF-mediated effects on GSIS, showing that both the protein and its protease activity are required for its action. CONCLUSIONS: We conclude that sPIF is a promising secretagogue for the treatment of T2D.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Insulisina , Ilhotas Pancreáticas , Camundongos , Humanos , Animais , Secreção de Insulina , Insulisina/metabolismo , Insulisina/farmacologia , Camundongos Obesos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Dieta Hiperlipídica/efeitos adversos , Ilhotas Pancreáticas/metabolismo
6.
Neuro Endocrinol Lett ; 44(4): 199-205, 2023 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-37466059

RESUMO

OBJECTIVE: It has been increasingly recognized that the pathological progress of Alzheimer´s disease (AD) is connected to metabolic function and inflammation. Insulin-degrading enzyme (IDE) is essential for glucose metabolism and the degradation of amyloid-ß. We aimed to explore the associations between IDE, total tau, and cytokines levels in plasma from subjects with AD and non-demented controls. METHODS AND MATERIAL: Plasma samples (18 patients diagnosed with AD and 6 non-demented controls) from the Netherlands Brain Bank were used to analyze IDE levels and total tau with an enzyme-linked immunosorbent assay. Cytokines were analyzed with Luminex custom plex assays for interleukin (IL)-6, IL-8, IL-10, and tumor necrosis factor-alpha (TNF-α). Results were analyzed using the Mann-Whitney U and Spearman´s rank correlation tests. RESULTS: Total tau in plasma was significantly increased in AD subjects compared to non-demented control subjects (p = 0.044). Total tau was positively correlated with IDE levels in plasma in all subjects (r = 0.494, p = 0.017). Significant correlations could be demonstrated between plasma levels of IDE and IL-6 (r = 0.546, p = 0.019), IL-8 (r = 0.664, p = 0.003), IL-10 (r = 0.833, p < 0.001), and TNF-α (r = 0.633, p = 0.005) in subjects with AD, but not in non-demented controls. CONCLUSION: Results from this study suggest that plasma IDE levels may be associated with inflammation and neurodegeneration and could potentially be a target for future diagnostic and treatment strategies.


Assuntos
Doença de Alzheimer , Insulisina , Humanos , Peptídeos beta-Amiloides , Citocinas , Inflamação , Insulisina/metabolismo , Interleucina-10 , Interleucina-6 , Interleucina-8 , Proteínas tau , Fator de Necrose Tumoral alfa
7.
Ageing Res Rev ; 90: 101999, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37414154

RESUMO

Accumulation of amyloid-ß in the central nervous system is a common feature of Alzheimer's disease (AD) and diabetes-related cognitive impairment. Since the insulin-degrading enzyme (IDE) can break down amyloid-ß plaques, there is considerable interest in using this enzyme to treat both neurological disorders. In this review, we have summarized the pre-clinical and clinical research on the potential application of IDE for the improvement of cognitive impairment. Furthermore, we have presented an overview of the main pathways that can be targeted to mitigate the progression of AD and the cognitive impairment caused by diabetes.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Diabetes Mellitus Tipo 2 , Insulisina , Humanos , Doença de Alzheimer/metabolismo , Insulisina/metabolismo , Peptídeos beta-Amiloides/metabolismo , Diabetes Mellitus Tipo 2/metabolismo
8.
Biomolecules ; 13(6)2023 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-37371470

RESUMO

Insulin-degrading enzyme (IDE) is a highly conserved metalloprotease that is mainly localized in the cytosol. Although IDE can degrade insulin and some other low molecular weight substrates efficiently, its ubiquitous expression suggests additional functions supported by experimental findings, such as a role in stress responses and cellular protein homeostasis. The translation of a long full-length IDE transcript has been reported to result in targeting to mitochondria, but the role of IDE in this compartment is unknown. To obtain initial leads on the function of IDE in mitochondria, we used a proximity biotinylation approach to identify proteins interacting with wild-type and protease-dead IDE targeted to the mitochondrial matrix. We find that IDE interacts with multiple mitochondrial ribosomal proteins as well as with proteins involved in the synthesis and assembly of mitochondrial complex I and IV. The mitochondrial interactomes of wild type and mutant IDE are highly similar and do not reveal any likely proteolytic IDE substrates. We speculate that IDE could adopt similar additional non-proteolytic functions in mitochondria as in the cytosol, acting as a chaperone and contributing to protein homeostasis and stress responses.


Assuntos
Transporte de Elétrons , Insulisina , Ribossomos Mitocondriais , Transporte de Elétrons/fisiologia , Insulina/metabolismo , Insulisina/metabolismo , Mitocôndrias/metabolismo , Ribossomos Mitocondriais/metabolismo , Peptídeo Hidrolases/metabolismo , Humanos
9.
Cell Mol Biol (Noisy-le-grand) ; 69(1): 13-18, 2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-37213163

RESUMO

Sertoli cells, the only somatic cells in testis seminiferous tubules, provide a supporting microenvironment for male germ cells and play essential roles in spermatogenesis. The insulin-degrading enzyme (IDE), a ubiquitous zinc peptidase of the inverzincin family, plays crucial role in sperm production, as IDE-knockout mice presented decreased testis weight and impaired sperm viability and morphology. However, whether and how IDE affects swine Sertoli cell proliferation remains unclear. Thus, in the present study, we aimed to evaluate the effects of IDE on the proliferation of swine Sertoli cells, as well as its underlying molecular mechanism. After knocking down IDE expression with small interfering RNA transfection, we analyzed the proliferation of swine Sertoli cells as well as the expression of related regulatory factors (WT1, ERK, and AKT). The results showed that IDE knockdown promoted swine Sertoli cell proliferation and increased WT1 expression, possibly through activating ERK and AKT. Overall, our findings suggest that IDE may be involved in male reproduction by regulating Sertoli cell proliferation, which provides new information to better understand the regulatory mechanisms of swine Sertoli cells and improve the reproductive traits of male pigs.


Assuntos
Insulisina , Células de Sertoli , Animais , Masculino , Proliferação de Células , Insulisina/genética , Insulisina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sêmen , Células de Sertoli/metabolismo , Suínos , Testículo/metabolismo
10.
Diabetologia ; 66(5): 884-896, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36884057

RESUMO

AIMS/HYPOTHESIS: Transcriptome analyses revealed insulin-gene-derived transcripts in non-beta endocrine islet cells. We studied alternative splicing of human INS mRNA in pancreatic islets. METHODS: Alternative splicing of insulin pre-mRNA was determined by PCR analysis performed on human islet RNA and single-cell RNA-seq analysis. Antisera were generated to detect insulin variants in human pancreatic tissue using immunohistochemistry, electron microscopy and single-cell western blot to confirm the expression of insulin variants. Cytotoxic T lymphocyte (CTL) activation was determined by MIP-1ß release. RESULTS: We identified an alternatively spliced INS product. This variant encodes the complete insulin signal peptide and B chain and an alternative C-terminus that largely overlaps with a previously identified defective ribosomal product of INS. Immunohistochemical analysis revealed that the translation product of this INS-derived splice transcript was detectable in somatostatin-producing delta cells but not in beta cells; this was confirmed by light and electron microscopy. Expression of this alternatively spliced INS product activated preproinsulin-specific CTLs in vitro. The exclusive presence of this alternatively spliced INS product in delta cells may be explained by its clearance from beta cells by insulin-degrading enzyme capturing its insulin B chain fragment and a lack of insulin-degrading enzyme expression in delta cells. CONCLUSIONS/INTERPRETATION: Our data demonstrate that delta cells can express an INS product derived from alternative splicing, containing both the diabetogenic insulin signal peptide and B chain, in their secretory granules. We propose that this alternative INS product may play a role in islet autoimmunity and pathology, as well as endocrine or paracrine function or islet development and endocrine destiny, and transdifferentiation between endocrine cells. INS promoter activity is not confined to beta cells and should be used with care when assigning beta cell identity and selectivity. DATA AVAILABILITY: The full EM dataset is available via www.nanotomy.org (for review: http://www.nanotomy.org/OA/Tienhoven2021SUB/6126-368/ ). Single-cell RNA-seq data was made available by Segerstolpe et al [13] and can be found at https://sandberglab.se/pancreas . The RNA and protein sequence of INS-splice was uploaded to GenBank (BankIt2546444 INS-splice OM489474).


Assuntos
Insulisina , Ilhotas Pancreáticas , Humanos , Células Secretoras de Somatostatina/metabolismo , Insulisina/metabolismo , Insulina/genética , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , RNA , Sinais Direcionadores de Proteínas
11.
Mol Nutr Food Res ; 67(7): e2200589, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36726048

RESUMO

SCOPE: Long-term high-fat diet (HFD) causes insulin resistance, which is a primary etiological factor in the development of obesity and type 2 diabetes mellitus. Impaired insulin clearance is not only a consequence but also a cause of insulin resistance. The kidney is a major site of insulin clearance, where the insulin-degrading enzyme (IDE) plays a vital role in the proximal tubule. Thus, the study investigates the role of renal IDE in the regulation of insulin resistance in HFD-induced obese mice. METHODS AND RESULTS: Twenty four-weeks of HFD in C57BL/6 mice causes insulin resistance and impaires insulin clearance, accompanied by a decrease in renal IDE expression and activity. Palmitic acid decreases IDE mRNA and protein expressions in HK-2 cells. RNA-Seq analysis found that the PPAR pathway is involved. 24-weeks of HFD decreases renal PPARγ, but not PPARα or PPARß/δ mRNA expression. The inhibition of IDE expression by palmitic acid is prevented by the PPARγ agonist rosiglitazone. The amount of PPARγ bound to the promoters of IDE is decreased in palmitic acid-treated cells. Rosiglitazone improves insulin clearance and insulin resistance and increases renal IDE expression in HFD fed-mice. CONCLUSION: Long-term HFD decreases renal IDE expression and activity, and causes insulin resistance, which involves PPARγ.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Insulisina , Camundongos , Animais , PPAR gama/genética , PPAR gama/metabolismo , Rosiglitazona , Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina/fisiologia , Insulisina/genética , Insulisina/metabolismo , Diabetes Mellitus Tipo 2/etiologia , Ácido Palmítico/farmacologia , Camundongos Endogâmicos C57BL , Insulina/metabolismo , Rim/metabolismo , Camundongos Obesos , RNA Mensageiro/metabolismo
12.
Neuromolecular Med ; 25(2): 193-204, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-35948857

RESUMO

Intercellular adhesion molecule 1 (ICAM1) is a vessel adhesion protein induced during brain vascular inflammation, which could be closely linked with the development of Alzheimer's disease (AD). This study investigated the effect of ICAM1 on amyloid-degrading enzymes (ADEs) in endothelial cells and their potential involvement in inflammation and AD progression. TNF-α treatment increased ICAM1 in human brain microvascular endothelial cells (HBMVECs) but decreased the neprilysin (NEP) protein level. Knock-down of ICAM1 using siRNA enhanced NEP, which increased the degradation of amyloid-ß. In the brains of 4-month-old AD transgenic mice (APPswe/PSEN1dE9), there were significantly higher levels of ICAM1 expression and amyloid deposits but lower levels of NEP and insulin-degrading enzymes (IDE), demonstrating an inverse correlation of ICAM1 with NEP and IDE expression. Further studies demonstrated significantly increased GFAP protein levels in the brain, specifically localized near blood vessels, of both TNF-α-injected and 4-month-old AD transgenic mice. Taken together, the induction of ICAM1 in endothelial cells suppresses NEP expression, accelerating the accumulation of amyloid-ß in blood vessels. It also enhances leukocyte adhesion to blood vessels stimulating the migration of leukocytes into the brain, subsequently triggering brain inflammation.


Assuntos
Doença de Alzheimer , Insulisina , Camundongos , Animais , Humanos , Lactente , Doença de Alzheimer/genética , Neprilisina/genética , Neprilisina/metabolismo , Neprilisina/farmacologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Molécula 1 de Adesão Intercelular/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Células Endoteliais/metabolismo , Peptídeos beta-Amiloides/metabolismo , Camundongos Transgênicos , Insulisina/genética , Insulisina/metabolismo , Insulisina/farmacologia , Encéfalo/metabolismo
13.
Biomolecules ; 12(12)2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36551314

RESUMO

Insulin degrading enzyme (IDE) has been detected in the cerebrospinal fluid media and plays a role in encapsulating and degrading the amyloid ß (Aß) monomer, thus regulating the levels of Aß monomers. The current work illustrates a first study by which IDE encapsulates polymorphic early-stage Aß oligomers. The main goal of this study was to investigate the molecular mechanisms of IDE activity on the encapsulated early-stage Aß dimers: fibril-like and random coil/α-helix dimers. Our work led to several findings. First, when the fibril-like Aß dimer interacts with IDE-C domain, IDE does not impede the contact between the monomers, but plays a role as a 'dead-end' chaperone protein. Second, when the fibril-like Aß dimer interacts with the IDE-N domain, IDE successfully impedes the contacts between monomers. Third, the inhibitory activity of IDE on random coil/α-helix dimers depends on the stability of the dimer. IDE could impede the contacts between monomers in relatively unstable random coil/α-helix dimers, but gets hard to impede in stable dimers. However, IDE encapsulates stable dimers and could serve as a 'dead-end' chaperone. Our results examine the molecular interactions between IDE and the dimers, and between the monomers within the dimers. Hence, this study provides insights into the inhibition mechanisms of the primary nucleation of Aß aggregation and the basic knowledge for rational design to inhibit Aß aggregation.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Insulisina , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Insulisina/metabolismo
14.
J Neurochem ; 163(6): 517-530, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36321194

RESUMO

Inflammation associated with viral infection of the nervous system has been involved in the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD) and multiple sclerosis. Polyinosinic:polycytidylic acid (poly[I:C]) is a Toll-like receptor 3 (TLR3) agonist that mimics the inflammatory response to systemic viral infections. Despite growing recognition of the role of glial cells in AD pathology, their involvement in the accumulation and clearance of amyloid ß (Aß) in the brain of patients with AD is poorly understood. Neprilysin (NEP) and insulin-degrading enzyme (IDE) are the main Aß-degrading enzymes in the brain. This study investigated whether poly(I:C) regulated Aß degradation and neurotoxicity by modulating NEP and IDE protein levels through TLR3 in astrocytes. To this aim, primary rat primary astrocyte cultures were treated with poly(I:C) and inhibitors of the TLR3 signaling. Protein levels were assessed by Western blot. Aß toxicity to primary neurons was measured by lactate dehydrogenase release. Poly(I:C) induced a significant decrease in NEP levels on the membrane of astrocytes as well as in the culture medium. The degradation of exogenous Aß was markedly delayed in poly(I:C)-treated astrocytes. This delay significantly increased the neurotoxicity of exogenous Aß1-42. Altogether, these results suggest that viral infections induce Aß neurotoxicity by decreasing NEP levels in astrocytes and consequently preventing Aß degradation.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Astrócitos , Insulisina , Neprilisina , Viroses , Animais , Ratos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/virologia , Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Astrócitos/virologia , Insulisina/metabolismo , Neprilisina/metabolismo , Receptor 3 Toll-Like/antagonistas & inibidores , Poli I-C/farmacologia , Viroses/complicações
15.
Sci Rep ; 12(1): 19808, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36396721

RESUMO

SIRT3 deacetylates mitochondrial proteins, thereby enhancing their function. We have previously demonstrated that Sirt3 gene deletion leads to brain mitochondrial dysfunction and neuroinflammation. We also reported that silencing of Sirt3 gene in APP/PS1 mice results in exacerbation of insulin resistance, neuroinflammation and ß amyloid plaque deposition. To further understand how metabolic syndrome and amyloid pathology interact, we performed RNA-seq analysis of the brain samples of APP/PS1/Sirt3-/- mice. Gene expression patterns were modulated in metabolic and inflammatory pathways by Sirt3 gene deletion, amyloid pathology, and the combination. Following Sirt3 gene deletion, a key finding was the decreased expression of insulin-degrading enzyme (IDE), an enzyme that regulates the levels of insulin and Aß peptides. Western diet feeding of Sirt3-/- and APP/PS1 mice resulted in decrease of IDE protein, parallel to Sirt3 downregulation. Conversely, activation of SIRT3 by nicotinamide riboside in vivo and in vitro resulted in IDE upregulation. SIRT3 activation in vivo also increased the levels of neprilysin, another Aß degrading enzyme and decreased the levels of BACE1 which generates Aß peptide suggesting SIRT3's role in amyloid plaque reduction. Our findings provide a plausible mechanism linking metabolic syndrome and amyloid pathology. SIRT3 may be a potential therapeutic target to treat AD.


Assuntos
Doença de Alzheimer , Insulisina , Síndrome Metabólica , Sirtuína 3 , Animais , Camundongos , Insulisina/genética , Insulisina/metabolismo , Sirtuína 3/genética , Sirtuína 3/metabolismo , Regulação para Baixo , Placa Amiloide , Doença de Alzheimer/metabolismo , Síndrome Metabólica/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Amiloide/metabolismo
16.
Int J Mol Sci ; 23(19)2022 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-36232381

RESUMO

Although the COVID-19 disease has developed into a worldwide pandemic, its pathophysiology remains to be fully understood. Insulin-degrading enzyme (IDE), a zinc-metalloprotease with a high affinity for insulin, has been found in the interactomes of multiple SARS-CoV-2 proteins. However, the relevance of IDE in the innate and adaptative immune responses elicited by circulating peripheral blood mononuclear cells is unknown. Here, we show that IDE is highly expressed on the surface of circulating monocytes, T-cells (both CD4+ and CD4-), and, to a lower extent, in B-cells from healthy controls. Notably, IDE's surface expression was upregulated on monocytes from COVID-19 patients at diagnosis, and it was increased in more severe patients. However, IDE's surface expression was downregulated (relative to healthy controls) 3 months after hospital discharge in all the studied immune subsets, with this effect being more pronounced in males than in females, and thus it was sex-dependent. Additionally, IDE levels in monocytes, CD4+ T-cells, and CD4- T-cells were inversely correlated with circulating insulin levels in COVID-19 patients (both at diagnosis and after hospital discharge). Of note, high glucose and insulin levels downregulated IDE surface expression by ~30% in the monocytes isolated from healthy donors, without affecting its expression in CD4+ T-cells and CD4- T-cells. In conclusion, our studies reveal the sex- and metabolism-dependent regulation of IDE in monocytes, suggesting that its regulation might be important for the recruitment of immune cells to the site of infection, as well as for glucometabolic control, in COVID-19 patients.


Assuntos
COVID-19 , Insulisina , Teste para COVID-19 , Feminino , Glucose , Hospitais , Humanos , Insulina/metabolismo , Insulisina/metabolismo , Leucócitos Mononucleares/metabolismo , Linfócitos/metabolismo , Masculino , Monócitos/metabolismo , SARS-CoV-2 , Zinco
17.
Neurochem Res ; 47(11): 3369-3384, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35915371

RESUMO

Previous studies have shown that ovarian estrogens are involved in the occurrence and pathology of Alzheimer's disease (AD) through regulation on hippocampal synaptic plasticity and spatial memory; however, the underlying mechanisms have not yet been elucidated at the genomic scale. In this study, we established the postmenopausal estrogen-deficient model by ovariectomy (OVX). Then, we used high-throughput Affymetrix Clariom transcriptomics and found 143 differentially expressed genes in the hippocampus of OVX mice with the absolute fold change ≥ 1.5 and P < 0.05. GO analysis showed that the highest enrichment was seen in long-term memory. Combined with the response to steroid hormone enrichment and GeneMANIA network prediction, the serum and glucocorticoid-regulated kinase 1 gene (Sgk1) was found to be the most potent candidate for ovarian estrogenic regulation. Sgk1 overexpression viral vectors (oSgk1) were then constructed and injected into the hippocampus of OVX mice. Morris water maze test revealed that the impaired spatial learning and memory induced by OVX was rescued by Sgk1 overexpression. Additionally, the altered expression of synaptic proteins and actin remodeling proteins and changes in CA1 spine density and synapse density induced by OVX were also significantly reversed by oSgk1. Moreover, the OVX-induced increase in Aß-producing BACE1 and Aß and the decrease in insulin degrading enzyme were significantly reversed by oSgk1. The above results show that multiple pathways and genes are involved in ovarian estrogenic regulation of the function of the hippocampus, among which Sgk1 may be a novel potent target against estrogen-sensitive hippocampal dysfunctions, such as Aß-initiated AD.


Assuntos
Doença de Alzheimer , Proteínas Imediatamente Precoces , Insulisina , Proteínas Serina-Treonina Quinases , Actinas/metabolismo , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Estrogênios/metabolismo , Feminino , Hipocampo/metabolismo , Proteínas Imediatamente Precoces/genética , Insulisina/metabolismo , Aprendizagem em Labirinto , Camundongos , Proteínas Serina-Treonina Quinases/genética , Aprendizagem Espacial , Memória Espacial/fisiologia , Transcriptoma
18.
Cells ; 11(16)2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-36010613

RESUMO

The evident implication of the insulin-degrading enzyme (IDE) in Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM), among its capacity to degrade insulin and amyloid-ß peptide (Aß), suggests that IDE could be an essential link in the relation between hyperinsulinemia, insulin resistance and AD. However, little is known about the cellular and molecular regulation of IDE expression, and even less has been explored regarding the post-transcriptional regulation of IDE, although it represents a great molecular target of interest for therapeutic treatments. We recently described that miR-7, a novel candidate for linking AD and T2DM at the molecular level, regulates IDE and other key genes in both pathologies, including some key genes involved in the insulin signaling pathway. Here, we explored whether other miRNAs as well as other post-transcriptional regulators, such as RNA binding proteins (RBP), could potentially participate in the regulation of IDE expression in vitro. Our data showed that in addition to miR-7, miR-125, miR-490 and miR-199 regulate IDE expression at the post-transcriptional level. Moreover, we also found that IDE contains multiple potential binding sites for several RBPs, and a narrow-down prediction analysis led us to speculate on a novel regulation of IDE by RALY and HuD. Taken together, these results demonstrate the novel players controlling IDE expression that could represent potential therapeutical targets to treat several metabolic diseases with a high impact on human health, including AD and T2DM.


Assuntos
Doença de Alzheimer , Diabetes Mellitus Tipo 2 , Insulisina , MicroRNAs , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo C , Humanos , Insulina/metabolismo , Insulisina/metabolismo , MicroRNAs/genética , MicroRNAs/uso terapêutico
19.
Int J Mol Sci ; 23(14)2022 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-35886965

RESUMO

The protozoan pathogen Cryptosporidium parvum infects intestinal epithelial cells and causes diarrhea in humans and young animals. Among the more than 20 genes encoding insulinase-like metalloproteinases (INS), two are paralogs with high sequence identity. In this study, one of them, INS-16 encoded by the cgd3_4270 gene, was expressed and characterized in a comparative study of its sibling, INS-15 encoded by the cgd3_4260 gene. A full-length INS-16 protein and its active domain I were expressed in Escherichia coli, and antibodies against the domain I and an INS-16-specific peptide were produced in rabbits. In the analysis of the crude extract of oocysts, a ~60 kDa fragment of INS-16 rather than the full protein was recognized by polyclonal antibodies against the specific peptide, indicating that INS-16 undergoes proteolytic cleavage before maturation. The expression of the ins-16 gene peaked at the invasion phase of in vitro C. parvum culture, with the documented expression of the protein in both sporozoites and merozoites. Localization studies with antibodies showed significant differences in the distribution of the native INS-15 and INS-16 proteins in sporozoites and merozoites. INS-16 was identified as a dense granule protein in sporozoites and macrogamonts but was mostly expressed at the apical end of merozoites. We screened 48 candidate INS-16 inhibitors from the molecular docking of INS-16. Among them, two inhibited the growth of C. parvum in vitro (EC50 = 1.058 µM and 2.089 µM). The results of this study suggest that INS-16 may have important roles in the development of C. parvum and could be a valid target for the development of effective treatments.


Assuntos
Cryptosporidium parvum , Insulisina , Metaloproteases , Proteínas de Protozoários , Animais , Criptosporidiose/metabolismo , Cryptosporidium/metabolismo , Cryptosporidium parvum/metabolismo , Insulisina/metabolismo , Metaloproteases/metabolismo , Simulação de Acoplamento Molecular , Proteínas de Protozoários/metabolismo , Coelhos , Esporozoítos/metabolismo
20.
Diabetologia ; 65(8): 1375-1389, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35652923

RESUMO

AIMS/HYPOTHESIS: Type 2 diabetes is characterised by hyperglucagonaemia and perturbed function of pancreatic glucagon-secreting alpha cells but the molecular mechanisms contributing to these phenotypes are poorly understood. Insulin-degrading enzyme (IDE) is present within all islet cells, mostly in alpha cells, in both mice and humans. Furthermore, IDE can degrade glucagon as well as insulin, suggesting that IDE may play an important role in alpha cell function in vivo. METHODS: We have generated and characterised a novel mouse model with alpha cell-specific deletion of Ide, the A-IDE-KO mouse line. Glucose metabolism and glucagon secretion in vivo was characterised; isolated islets were tested for glucagon and insulin secretion; alpha cell mass, alpha cell proliferation and α-synuclein levels were determined in pancreas sections by immunostaining. RESULTS: Targeted deletion of Ide exclusively in alpha cells triggers hyperglucagonaemia and alpha cell hyperplasia, resulting in elevated constitutive glucagon secretion. The hyperglucagonaemia is attributable in part to dysregulation of glucagon secretion, specifically an impaired ability of IDE-deficient alpha cells to suppress glucagon release in the presence of high glucose or insulin. IDE deficiency also leads to α-synuclein aggregation in alpha cells, which may contribute to impaired glucagon secretion via cytoskeletal dysfunction. We showed further that IDE deficiency triggers impairments in cilia formation, inducing alpha cell hyperplasia and possibly also contributing to dysregulated glucagon secretion and hyperglucagonaemia. CONCLUSIONS/INTERPRETATION: We propose that loss of IDE function in alpha cells contributes to hyperglucagonaemia in type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Glucagon , Células Secretoras de Insulina , Insulisina , Animais , Proliferação de Células/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucagon/metabolismo , Células Secretoras de Glucagon/metabolismo , Hiperplasia/genética , Hiperplasia/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Insulisina/genética , Insulisina/metabolismo , Camundongos , alfa-Sinucleína/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...