Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
J Biol Chem ; 299(5): 104682, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37030503

RESUMO

Islet amyloid polypeptide (amylin) secreted from the pancreas crosses from the blood to the brain parenchyma and forms cerebral mixed amylin-ß amyloid (Aß) plaques in persons with Alzheimer's disease (AD). Cerebral amylin-Aß plaques are found in both sporadic and early-onset familial AD; however, the role of amylin-Aß co-aggregation in potential mechanisms underlying this association remains unknown, in part due to lack of assays for detection of these complexes. Here, we report the development of an ELISA to detect amylin-Aß hetero-oligomers in brain tissue and blood. The amylin-Aß ELISA relies on a monoclonal anti-Aß mid-domain antibody (detection) and a polyclonal anti-amylin antibody (capture) designed to recognize an epitope that is distinct from the high affinity amylin-Aß binding sites. The utility of this assay is supported by the analysis of molecular amylin-Aß codeposition in postmortem brain tissue obtained from persons with and without AD pathology. By using transgenic AD-model rats, we show that this new assay can detect circulating amylin-Aß hetero-oligomers in the blood and is sensitive to their dissociation to monomers. This is important because therapeutic strategies to block amylin-Aß co-aggregation could reduce or delay the development and progression of AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Encéfalo , Animais , Camundongos , Ratos , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/genética , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Camundongos Transgênicos , Pâncreas/metabolismo , Ratos Transgênicos
2.
J Stroke Cerebrovasc Dis ; 32(9): 107300, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37572602

RESUMO

BACKGROUND AND PURPOSE: Human amylin is a 37 amino-acid pancreatic peptide that forms neuro-toxic aggregates that deposit in the endothelium of brain capillaries of patients with diabetes, potentially contributing to cerebral small vessel ischemic injury. Pathogenic amylin also deposits in the capillary endothelium in other organs, including the skin. The aim of this study was to test the hypothesis that skin capillary amylin deposition correlates with cerebral small vessel amylin deposition, potentially providing a clinically useful marker of cerebral amylin deposition. METHODS: Immunohistochemistry (IHC) was performed for human amylin and collagen IV in brain and skin sections of rats (age 15-16 months) with pancreatic overexpression of amyloidogenic human amylin polypeptide (HIP rats), and control rats (Wild type; WT; rats that express non-amyloidogenic rat amylin) using antibodies binding amylin (n = 5 male and 5 female rats for each group) and antibodies binding Hypoxia inducing factor (HIF)-1α and HIF-2α (n = 3 for each group). The reactive amylin-aldehyde 4-hydroxynonenal (4-HNE) adduct was measured in skin homogenates. (n = 4 for each group) RESULTS: Brain capillaries isolated from HIP rats had higher amylin content compared to WT rats using Western blot with anti-amylin antibody (p = 0.0010). The HIF-1α and HIF-2α immunoreactivity signals in skin from HIP and WT rats were similar (p = 0.2 for HIF-1 α, and p = 0.75 for HIF-2α). Amylin-4HNE adduct formation was higher in HIP rats compared to WT rats (p = 0.0014). There was phenotypic similarity between brain and skin capillary amylin based on co-staining for human amylin and collagen IV in both HIP and WT rats. CONCLUSION: Skin and brain capillary amylin deposition are similar providing evidence that a skin biopsy might be providing a potential biomarker for diabetes-associated intracranial vasculopathy.


Assuntos
Capilares , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Ratos , Humanos , Masculino , Animais , Feminino , Lactente , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Capilares/metabolismo , Encéfalo/patologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Colágeno/metabolismo
3.
Stroke ; 52(6): e244-e249, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33947210

RESUMO

Recent histological analyses of human brains show that small vessel-type injuries in the setting of type-2 diabetes colocalize with deposits of amylin, an amyloid-forming hormone secreted by the pancreas. Amylin inclusions are also identified in circulating red blood cells in people with type-2 diabetes and stroke or cardiovascular disease. In laboratory models of type-2 diabetes, accumulation of aggregated amylin in blood and the cerebral microvasculature induces brain microhemorrhages and reduces cerebral blood flow leading to white matter ischemia and neurological deficits. At the cellular level, aggregated amylin causes cell membrane lipid peroxidation injury, downregulation of tight junction proteins, and activation of proinflammatory signaling pathways which, in turn, induces macrophage activation and macrophage infiltration in vascular areas positive for amylin deposition. We review each step of this cascade based on experimental and clinical evidence and propose the hypothesis that systemic amylin dyshomeostasis may underlie the disparity between glycemic control and stroke risk and may be a therapeutic target to reduce the risk of small vessel ischemic stroke in patients with type-2 diabetes.


Assuntos
Doenças de Pequenos Vasos Cerebrais/sangue , Diabetes Mellitus Tipo 2/sangue , AVC Isquêmico/sangue , Polipeptídeo Amiloide das Ilhotas Pancreáticas/sangue , Biomarcadores/sangue , Doenças de Pequenos Vasos Cerebrais/etiologia , Diabetes Mellitus Tipo 2/complicações , Humanos , AVC Isquêmico/etiologia
4.
Kidney Int ; 97(1): 143-155, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31739987

RESUMO

In the setting of type-2 diabetes, there are declines of structural stability and functionality of blood capillaries and red blood cells (RBCs), increasing the risk for microcirculatory disturbances. Correcting hyperglycemia is not entirely effective at reestablishing normal cellular metabolism and function. Therefore, identification of pathological changes occurring before the development of overt hyperglycemia may lead to novel therapeutic targets for reducing the risk of microvascular dysfunction. Here we determine whether RBC-capillary interactions are altered by prediabetic hypersecretion of amylin, an amyloid forming hormone co-synthesized with insulin, and is reversed by endothelial cell-secreted epoxyeicosatrienoic acids. In patients, we found amylin deposition in RBCs in association with type-2 diabetes, heart failure, cancer and stroke. Amylin-coated RBCs have altered shape and reduced functional (non-glycated) hemoglobin. Amylin-coated RBCs administered intravenously in control rats upregulated erythropoietin and renal arginase expression and activity. We also found that diabetic rats expressing amyloid-forming human amylin in the pancreas (the HIP rat model) have increased tissue levels of hypoxia-inducible transcription factors, compared to diabetic rats that express non-amyloid forming rat amylin (the UCD rat model). Upregulation of erythropoietin correlated with lower hematocrit in the HIP model indicating pathologic erythropoiesis. In the HIP model, pharmacological upregulation of endogenous epoxyeicosatrienoic acids protected the renal microvasculature against amylin deposition and also reduced renal accumulation of HIFs. Thus, prediabetes induces dysregulation of amylin homeostasis and promotes amylin deposition in RBCs and the microvasculature altering RBC-capillary interaction leading to activation of hypoxia signaling pathways and pathologic erythropoiesis. Hence, dysregulation of amylin homeostasis could be a therapeutic target for ameliorating diabetic vascular complications.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Angiopatias Diabéticas/patologia , Eritrócitos/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Microvasos/patologia , Adulto , Amiloide/metabolismo , Animais , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/induzido quimicamente , Diabetes Mellitus Tipo 2/genética , Angiopatias Diabéticas/sangue , Modelos Animais de Doenças , Eicosanoides/metabolismo , Eritropoese , Eritropoetina/metabolismo , Feminino , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Rim/irrigação sanguínea , Rim/patologia , Masculino , Microcirculação , Pessoa de Meia-Idade , Ratos , Estudos Retrospectivos
5.
Nature ; 502(7471): 372-6, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24077098

RESUMO

Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is an enzyme with important regulatory functions in the heart and brain, and its chronic activation can be pathological. CaMKII activation is seen in heart failure, and can directly induce pathological changes in ion channels, Ca(2+) handling and gene transcription. Here, in human, rat and mouse, we identify a novel mechanism linking CaMKII and hyperglycaemic signalling in diabetes mellitus, which is a key risk factor for heart and neurodegenerative diseases. Acute hyperglycaemia causes covalent modification of CaMKII by O-linked N-acetylglucosamine (O-GlcNAc). O-GlcNAc modification of CaMKII at Ser 279 activates CaMKII autonomously, creating molecular memory even after Ca(2+) concentration declines. O-GlcNAc-modified CaMKII is increased in the heart and brain of diabetic humans and rats. In cardiomyocytes, increased glucose concentration significantly enhances CaMKII-dependent activation of spontaneous sarcoplasmic reticulum Ca(2+) release events that can contribute to cardiac mechanical dysfunction and arrhythmias. These effects were prevented by pharmacological inhibition of O-GlcNAc signalling or genetic ablation of CaMKIIδ. In intact perfused hearts, arrhythmias were aggravated by increased glucose concentration through O-GlcNAc- and CaMKII-dependent pathways. In diabetic animals, acute blockade of O-GlcNAc inhibited arrhythmogenesis. Thus, O-GlcNAc modification of CaMKII is a novel signalling event in pathways that may contribute critically to cardiac and neuronal pathophysiology in diabetes and other diseases.


Assuntos
Arritmias Cardíacas/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Complicações do Diabetes/metabolismo , Hiperglicemia/metabolismo , Acetilglucosamina/metabolismo , Animais , Arritmias Cardíacas/complicações , Arritmias Cardíacas/enzimologia , Benzilaminas/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Cálcio/metabolismo , Complicações do Diabetes/enzimologia , Diazo-Oxo-Norleucina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Glucose/metabolismo , Glucose/farmacologia , Glicosilação/efeitos dos fármacos , Humanos , Hiperglicemia/complicações , Hiperglicemia/enzimologia , Camundongos , Miocárdio/citologia , Miocárdio/enzimologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , Ratos , Retículo Sarcoplasmático/metabolismo , Sulfonamidas/farmacologia
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt B): 1923-1930, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29066284

RESUMO

Amylin is a pancreatic ß-cell hormone co-secreted with insulin, plays a role in normal glucose homeostasis, and forms amyloid in the pancreatic islets of individuals with type-2 diabetes. Aggregated amylin is also found in blood and extra-pancreatic tissues, including myocardium. Myocardial amylin accumulation is associated with myocyte Ca2+ dysregulation in diabetic rats expressing human amylin. Whether deposition of amylin in the heart is a consequence of or a contributor to diabetic cardiomyopathy remains unknown. We used amylin knockout (AKO) mice intravenously infused with either human amylin (i.e, the aggregated form) or non-amyloidogenic (i.e., monomeric) rodent amylin to test the hypothesis that aggregated amylin accumulates in the heart in the absence of diabetes. AKO mice infused with human amylin, but not rodent amylin, showed amylin deposits in the myocardium. Cardiac amylin level was larger in males compared to females. Sarcolemmal Ca2+ leak and Ca2+ transients were increased in myocytes isolated from males infused with human amylin while no significant changes occurred in either females injected with human amylin or in rat amylin-infused mice. In isolated cardiac myocytes, the amylin receptor antagonist AC-187 did not effectively block the interaction of amylin with the sarcolemma. In conclusion, circulating aggregated amylin accumulates preferentially in male vs. female hearts and its effects on myocyte Ca2+ cycling do not require diabetic remodeling of the myocardium. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Cardiomiopatias Diabéticas/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Miócitos Cardíacos/metabolismo , Sarcolema/metabolismo , Remodelação Ventricular , Animais , Cardiomiopatias Diabéticas/genética , Cardiomiopatias Diabéticas/patologia , Cardiomiopatias Diabéticas/fisiopatologia , Modelos Animais de Doenças , Feminino , Mediadores da Inflamação/sangue , Infusões Intravenosas , Interleucina-1beta/sangue , Polipeptídeo Amiloide das Ilhotas Pancreáticas/administração & dosagem , Polipeptídeo Amiloide das Ilhotas Pancreáticas/deficiência , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Masculino , Camundongos Knockout , Miócitos Cardíacos/patologia , Agregados Proteicos , Agregação Patológica de Proteínas , Sarcolema/patologia , Fatores Sexuais
7.
Ann Neurol ; 82(2): 208-222, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28696548

RESUMO

OBJECTIVE: The brain blood vessels of patients with type 2 diabetes and dementia have deposition of amylin, an amyloidogenic hormone cosecreted with insulin. It is not known whether vascular amylin deposition is a consequence or a trigger of vascular injury. We tested the hypothesis that the vascular amylin deposits cause endothelial dysfunction and microvascular injury and are modulated by amylin transport in the brain via plasma apolipoproteins. METHODS: Rats overexpressing amyloidogenic (human) amylin in the pancreas (HIP rats) and amylin knockout (AKO) rats intravenously infused with aggregated amylin were used for in vivo phenotyping. We also carried out biochemical analyses of human brain tissues and studied the effects of the aggregated amylin on endothelial cells ex vivo. RESULTS: Amylin deposition in brain blood vessels is associated with vessel wall disruption and abnormal surrounding neuropil in patients with type 2 diabetes and dementia, in HIP rats, and in AKO rats infused with aggregated amylin. HIP rats have brain microhemorrhages, white matter injury, and neurologic deficits. Vascular amylin deposition provokes loss of endothelial cell coverage and tight junctions. Intravenous infusion in AKO rats of human amylin, or combined human amylin and apolipoprotein E4, showed that amylin binds to plasma apolipoproteins. The intravenous infusion of apolipoprotein E4 exacerbated the brain accumulation of aggregated amylin and vascular pathology in HIP rats. INTERPRETATION: These data identify vascular amylin deposition as a trigger of brain endothelial dysfunction that is modulated by plasma apolipoproteins and represents a potential therapeutic target in diabetes-associated dementia and stroke. Ann Neurol 2017;82:208-222.


Assuntos
Encéfalo/patologia , Diabetes Mellitus Tipo 2/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/efeitos adversos , Leucoencefalopatias/induzido quimicamente , Leucoencefalopatias/patologia , Microvasos/metabolismo , Idoso de 80 Anos ou mais , Animais , Apolipoproteína E4/administração & dosagem , Apolipoproteína E4/efeitos adversos , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Sinergismo Farmacológico , Endotélio/metabolismo , Técnicas de Inativação de Genes , Humanos , Hemorragias Intracranianas/induzido quimicamente , Polipeptídeo Amiloide das Ilhotas Pancreáticas/sangue , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Leucoencefalopatias/sangue , Leucoencefalopatias/complicações , Imageamento por Ressonância Magnética , Aprendizagem em Labirinto/efeitos dos fármacos , Destreza Motora/efeitos dos fármacos , Neuroimagem , Pâncreas/metabolismo , Ratos , Ratos Mutantes , Junções Íntimas/efeitos dos fármacos
9.
Expert Rev Proteomics ; 12(6): 575-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26503000

RESUMO

Type II diabetes increases the risk for cognitive decline via multiple traits. Amylin is a pancreatic hormone that has amyloidogenic and cytotoxic properties similar to the amyloid-ß peptide. The amylin hormone is overexpressed in individuals with pre-diabetic insulin resistance or obesity leading to amylin oligomerization and deposition in pancreatic islets. Amylin oligomerization was implicated in the apoptosis of the insulin-producing ß-cells. Recent studies showed that brain tissue from diabetic patients with cerebrovascular dementia or Alzheimer's disease contains significant deposits of oligomerized amylin. It has also been reported that the brain amylin deposition reduced exploratory drive, recognition memory and vestibulomotor function in a rat model that overexpresses human amylin in the pancreas. These novel findings are reviewed here and the hypothesis that type II diabetes is linked with cognitive decline by amylin accumulation in the brain is proposed. Deciphering the impact of hyperamylinemia on the brain is critical for both etiology and treatment of dementia.


Assuntos
Demência/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Animais , Demência/etiologia , Diabetes Mellitus Tipo 2/complicações , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética
10.
Ann Neurol ; 74(4): 517-26, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23794448

RESUMO

OBJECTIVE: Hyperamylinemia, a common pancreatic disorder in obese and insulin-resistant patients, is known to cause amylin oligomerization and cytotoxicity in pancreatic islets, leading to ß-cell mass depletion and development of type 2 diabetes. Recent data has revealed that hyperamylinemia also affects the vascular system, heart, and kidneys. We therefore hypothesized that oligomerized amylin might accumulate in the cerebrovascular system and brain parenchyma of diabetic patients. METHODS: Amylin accumulation in the brain of diabetic patients with vascular dementia or Alzheimer disease (AD), nondiabetic patients with AD, and age-matched healthy controls was assessed by quantitative real time polymerase chain reaction, immunohistochemistry, Western blot, and enzyme-linked immunosorbent assay. RESULTS: Amylin oligomers and plaques were identified in the temporal lobe gray matter from diabetic patients, but not controls. In addition, extensive amylin deposition was found in blood vessels and perivascular spaces. Intriguingly, amylin deposition was also detected in blood vessels and brain parenchyma of patients with late onset AD without clinically apparent diabetes. Mixed amylin and amyloid ß (Aß) deposits were occasionally observed. However, amylin accumulation leads to amyloid formation independent of Aß deposition. Tissues infiltrated by amylin showed increased interstitial space, vacuolation, spongiform change, and capillaries bent at amylin accumulation sites. Unlike the pancreas, there was no evidence of amylin synthesis in the brain. INTERPRETATION: Metabolic disorders and aging promote accumulation of amylin amyloid in the cerebrovascular system and gray matter, altering microvasculature and tissue structure. Amylin amyloid formation in the wall of cerebral blood vessels may also induce failure of elimination of Aß from the brain, thus contributing to the etiology of AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Angiopatias Diabéticas/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/genética , Feminino , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Masculino , RNA Mensageiro/metabolismo
11.
Circ Res ; 110(4): 598-608, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22275486

RESUMO

RATIONALE: Hyperamylinemia is common in patients with obesity and insulin resistance, coincides with hyperinsulinemia, and results in amyloid deposition. Amylin amyloids are generally considered a pancreatic disorder in type 2 diabetes. However, elevated circulating levels of amylin may also lead to amylin accumulation and proteotoxicity in peripheral organs, including the heart. OBJECTIVE: To test whether amylin accumulates in the heart of obese and type 2 diabetic patients and to uncover the effects of amylin accumulation on cardiac morphology and function. METHODS AND RESULTS: We compared amylin deposition in failing and nonfailing hearts from lean, obese, and type 2 diabetic humans using immunohistochemistry and Western blots. We found significant accumulation of large amylin oligomers, fibrils, and plaques in failing hearts from obese and diabetic patients but not in normal hearts and failing hearts from lean, nondiabetic humans. Small amylin oligomers were even elevated in nonfailing hearts from overweight/obese patients, suggesting an early state of accumulation. Using a rat model of hyperamylinemia transgenic for human amylin, we observed that amylin oligomers attach to the sarcolemma, leading to myocyte Ca(2+) dysregulation, pathological myocyte remodeling, and diastolic dysfunction, starting from prediabetes. In contrast, prediabetic rats expressing the same level of wild-type rat amylin, a nonamyloidogenic isoform, exhibited normal heart structure and function. CONCLUSIONS: Hyperamylinemia promotes amylin deposition in the heart, causing alterations of cardiac myocyte structure and function. We propose that detection and disruption of cardiac amylin buildup may be both a predictor of heart dysfunction and a novel therapeutic strategy in diabetic cardiomyopathy.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Cardiomiopatias Diabéticas/etiologia , Insuficiência Cardíaca/etiologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Miocárdio/metabolismo , Obesidade/complicações , Estado Pré-Diabético/complicações , Adulto , Idoso , Animais , Western Blotting , Sinalização do Cálcio , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatias Diabéticas/diagnóstico por imagem , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/fisiopatologia , Modelos Animais de Doenças , Feminino , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Histona Desacetilases/metabolismo , Humanos , Imuno-Histoquímica , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Masculino , Pessoa de Meia-Idade , Miocárdio/patologia , Fatores de Transcrição NFATC/metabolismo , Obesidade/metabolismo , Estado Pré-Diabético/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Ultrassonografia , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda , Pressão Ventricular , Remodelação Ventricular , Adulto Jovem
12.
Biophys Chem ; 310: 107252, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38663120

RESUMO

Amylin is a systemic neuroendocrine hormone co-expressed and co-secreted with insulin by pancreatic ß-cells. In persons with thype-2 diabetes, amylin forms pancreatic amyloid triggering inflammasome and interleukin-1ß signaling and inducing ß-cell apoptosis. Here, we summarize recent progress in understanding the potential link between amyloid-forming pancreatic amylin and Alzheimer's disease (AD). Clinical data describing amylin pathology in AD alongside mechanistic studies in animals are reviewed. Data from multiple research teams indicate higher amylin concentrations are associated with increased frequency of cognitive impairment and amylin co-aggregates with ß-amyloid in AD-type dementia. Evidence from rodent models further suggests cerebrovascular amylin accumulation as a causative factor underlying neurological deficits. Analysis of relevant literature suggests that modulating the amylin-interleukin-1ß pathway may provide an approach for counteracting neuroinflammation in AD.


Assuntos
Doença de Alzheimer , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Doenças Neuroinflamatórias , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Humanos , Animais , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Inflamassomos/metabolismo
13.
Curr Alzheimer Res ; 2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36803745

RESUMO

Alzheimer Disease (AD) pathology has been linked to brain accumulation of ß amyloid (Aß) and neurofibrillary tau tangles. An intriguing question is whether targeting therapeutically factors independent of Aß and tau pathologies could delay or even stop neurodegeneration. Amylin, a pancreatic hormone co-secreted with insulin, is believed to play a role in the central regulation of satiation and was shown to form pancreatic amyloid in persons with type-2 diabetes mellitus. Accumulating evidence demonstrates that amyloid-forming amylin secreted from the pancreas synergistically aggregates with vascular and parenchymal Aß in the brain, in both sporadic and early-onset familial AD. Pancreatic expression of amyloid-forming human amylin in AD-model rats accelerates AD-like pathology, whereas genetically suppressed amylin secretion protects against AD effects. Thus, current data suggest a role of pancreatic amyloid-forming amylin in modifying AD; further research is required to test whether lowering circulating amylin levels early during AD pathogenesis may curb cognitive decline.

14.
Front Endocrinol (Lausanne) ; 14: 1104662, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36875454

RESUMO

Chronic kidney disease (CKD) is increasing worldwide and is associated with diabetic states (obesity, prediabetes and type-2 diabetes mellitus). The kidney is intrinsically susceptible to low oxygen (hypoxia) and renal hypoxia plays a vital role in the progression of CKD. Recent studies suggest an association between CKD and renal deposition of amyloid-forming amylin secreted from the pancreas. Renal accumulation of amyloid-forming amylin is associated with hypertension, mitochondrial dysfunction, increased production of reactive oxygen species (ROS) and activation of hypoxia signaling in the kidney. In this review we will discuss potential associations between renal amylin amyloid accumulation, hypertension, and mechanism of hypoxia-induced kidney dysfunction, including activation of hypoxia-inducible factors (HIFs) and mitochondrial dysfunction.


Assuntos
Hipertensão , Hormônios Peptídicos , Insuficiência Renal Crônica , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Proteínas Amiloidogênicas , Rim , Isquemia , Hipóxia
15.
Mol Nutr Food Res ; : e2200405, 2023 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-36708219

RESUMO

Type-2 diabetes raises the risk for Alzheimer's disease (AD)-type dementia and the conversion from mild cognitive impairment to dementia, yet mechanisms connecting type-2 diabetes to AD remain largely unknown. Amylin, a pancreatic ß-cell hormone co-secreted with insulin, participates in the central regulation of satiation, but also forms pancreatic amyloid in persons with type-2 diabetes and synergistically interacts with brain amyloid ß (Aß) pathology, in both sporadic and familial Alzheimer's disease (AD). Growing evidence from studies of tumor growth, together with early observations in skeletal muscle, indicates amylin as a potential trigger of cellular metabolic reprogramming. Because the blood, cerebrospinal fluid, and brain parenchyma in humans with AD have increased concentrations of amylin, amylin-mediated pathological processes in the brain may involve neuronal metabolic remodeling. This review summarizes recent progress in understanding the link between prediabetic hypersecretion of amylin and risk of neuronal metabolic remodeling and AD and suggests nutritional and medical effects of food constituents that might prevent and/or ameliorate amylin-mediated neuronal metabolic remodeling.

16.
J Am Heart Assoc ; 12(14): e029997, 2023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37421267

RESUMO

Background Mitochondrial dysfunction contributes to the cardiac remodeling triggered by type 2 diabetes (T2D). Mitochondrial Ca2+ concentration ([Ca2+]m) modulates the oxidative state and cytosolic Ca2+ regulation. Thus, we investigated how T2D affects mitochondrial Ca2+ fluxes, the downstream consequences on myocyte function, and the effects of normalizing mitochondrial Ca2+ transport. Methods and Results We compared myocytes/hearts from transgenic rats with late-onset T2D (rats that develop late-onset T2D due to heterozygous expression of human amylin in the pancreatic ß-cells [HIP] model) and their nondiabetic wild-type (WT) littermates. [Ca2+]m was significantly lower in myocytes from diabetic HIP rats compared with WT cells. Ca2+ extrusion through the mitochondrial Na+/Ca2+ exchanger (mitoNCX) was elevated in HIP versus WT myocytes, particularly at moderate and high [Ca2+]m, while mitochondrial Ca2+ uptake was diminished. Mitochondrial Na+ concentration was comparable in WT and HIP rat myocytes and remained remarkably stable while manipulating mitoNCX activity. Lower [Ca2+]m was associated with oxidative stress, increased sarcoplasmic reticulum Ca2+ leak in the form of Ca2+ sparks, and mitochondrial dysfunction in T2D hearts. MitoNCX inhibition with CGP-37157 reduced oxidative stress, Ca2+ spark frequency, and stress-induced arrhythmias in HIP rat hearts while having no significant effect in WT rats. In contrast, activation of the mitochondrial Ca2+ uniporter with SB-202190 enhanced spontaneous sarcoplasmic reticulum Ca2+ release and had no significant effect on arrhythmias in both WT and HIP rat hearts. Conclusions [Ca2+]m is reduced in myocytes from rats with T2D due to a combination of exacerbated mitochondrial Ca2+ extrusion through mitoNCX and impaired mitochondrial Ca2+ uptake. Partial mitoNCX inhibition limits sarcoplasmic reticulum Ca2+ leak and arrhythmias in T2D hearts, whereas mitochondrial Ca2+ uniporter activation does not.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Ratos , Humanos , Animais , Miócitos Cardíacos/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Diabetes Mellitus Experimental/metabolismo , Arritmias Cardíacas/metabolismo , Retículo Sarcoplasmático/metabolismo , Sinalização do Cálcio/fisiologia , Mitocôndrias/metabolismo , Cálcio/metabolismo
17.
Commun Biol ; 6(1): 2, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36596993

RESUMO

Impairment of vascular pathways of cerebral ß-amyloid (Aß) elimination contributes to Alzheimer disease (AD). Vascular damage is commonly associated with diabetes. Here we show in human tissues and AD-model rats that bloodborne islet amyloid polypeptide (amylin) secreted from the pancreas perturbs cerebral Aß clearance. Blood amylin concentrations are higher in AD than in cognitively unaffected persons. Amyloid-forming amylin accumulates in circulating monocytes and co-deposits with Aß within the brain microvasculature, possibly involving inflammation. In rats, pancreatic expression of amyloid-forming human amylin indeed induces cerebrovascular inflammation and amylin-Aß co-deposits. LRP1-mediated Aß transport across the blood-brain barrier and Aß clearance through interstitial fluid drainage along vascular walls are impaired, as indicated by Aß deposition in perivascular spaces. At the molecular level, cerebrovascular amylin deposits alter immune and hypoxia-related brain gene expression. These converging data from humans and laboratory animals suggest that altering bloodborne amylin could potentially reduce cerebrovascular amylin deposits and Aß pathology.


Assuntos
Doença de Alzheimer , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Humanos , Ratos , Animais , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteínas Amiloidogênicas , Pâncreas/metabolismo , Inflamação
18.
Am J Physiol Heart Circ Physiol ; 303(7): H853-62, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22865388

RESUMO

Glycemic regulation improves myocardial function in diabetic patients, but finding optimal therapeutic strategies remains challenging. Recent data have shown that pharmacological inhibition of soluble epoxide hydrolase (sEH), an enzyme that decreases the endogenous levels of protective epoxyeicosatrienoic acids (EETs), improves glucose homeostasis in insulin-resistant mice. Here, we tested whether the administration of sEH inhibitors preserves cardiac myocyte structure and function in hyperglycemic rats. University of California-Davis-type 2 diabetes mellitus (UCD-T2DM) rats with nonfasting blood glucose levels in the range of 150-200 mg/dl were treated with the sEH inhibitor 1-(1-acetypiperidin-4-yl)-3-adamantanylurea (APAU) for 6 wk. Administration of APAU attenuated the progressive increase of blood glucose concentration and preserved mitochondrial structure and myofibril morphology in cardiac myocytes, as revealed by electron microscopy imaging. Fluorescence microscopy with Ca(2+) indicators also showed a 40% improvement of cardiac Ca(2+) transients in treated rats. Sarcoplasmic reticulum Ca(2+) content was decreased in both treated and untreated rats compared with control rats. However, treatment limited this reduction by 30%, suggesting that APAU may protect the intracellular Ca(2+) effector system. Using Western blot analysis on cardiac myocyte lysates, we found less downregulation of sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA), the main route of Ca(2+) reuptake in the sarcoplasmic reticulum, and lower expression of hypertrophic markers in treated versus untreated UCD-T2DM rats. In conclusion, APAU enhances the therapeutic effects of EETs, resulting in slower progression of hyperglycemia, efficient protection of myocyte structure, and reduced Ca(2+) dysregulation and SERCA remodeling in hyperglycemic rats. The results suggest that sEH/EETs may be an effective therapeutic target for cardioprotection in insulin resistance and diabetes.


Assuntos
Adamantano/análogos & derivados , Complicações do Diabetes/prevenção & controle , Diabetes Mellitus Tipo 2/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/antagonistas & inibidores , Cardiopatias/prevenção & controle , Hipoglicemiantes/uso terapêutico , Miócitos Cardíacos/efeitos dos fármacos , Ureia/análogos & derivados , Adamantano/farmacologia , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Western Blotting , Sinalização do Cálcio/efeitos dos fármacos , Cruzamentos Genéticos , Complicações do Diabetes/sangue , Complicações do Diabetes/enzimologia , Complicações do Diabetes/etiologia , Complicações do Diabetes/patologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/enzimologia , Modelos Animais de Doenças , Progressão da Doença , Eicosanoides/metabolismo , Epóxido Hidrolases/metabolismo , Cardiopatias/sangue , Cardiopatias/enzimologia , Cardiopatias/etiologia , Cardiopatias/patologia , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/ultraestrutura , Miofibrilas/efeitos dos fármacos , Miofibrilas/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Fatores de Tempo , Ureia/farmacologia
19.
Sci Rep ; 11(1): 20926, 2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34686739

RESUMO

Population-based studies identified an association between a prior pregnancy complicated by gestational diabetes mellitus (GDM) and cardiac hypertrophy and dysfunction later in life. It is however unclear whether GDM initiates this phenotype and what are the underlying mechanisms. We addressed these questions by using female rats that express human amylin (HIP rats) as a GDM model and their wild-type (WT) littermates as the normal pregnancy model. Pregnant and two months postpartum HIP females had increased left-ventricular mass and wall thickness compared to non-pregnant HIP females, which indicates the presence of concentric hypertrophy. These parameters were unchanged in WT females during both pregnancy and postpartum periods. Hypertrophic Ca2+-dependent calcineurin/NFAT signaling was stimulated two months after giving birth in HIP females but not in the WT. In contrast, the CaMKII/HDAC hypertrophy pathway was active immediately after giving birth and returned to the baseline by two months postpartum in both WT and HIP females. Myocytes from two months postpartum HIP females exhibited slower Ca2+ transient relaxation and higher diastolic Ca2+ levels, which may explain calcineurin activation. No such effects occurred in the WT. These results suggest that a GDM-complicated pregnancy accelerates the development of pathological cardiac remodeling likely through activation of calcineurin/NFAT signaling.


Assuntos
Calcineurina/metabolismo , Cardiomegalia/metabolismo , Diabetes Gestacional/metabolismo , Fatores de Transcrição NFATC/metabolismo , Período Pós-Parto/metabolismo , Transdução de Sinais/fisiologia , Animais , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Feminino , Miócitos Cardíacos/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Remodelação Ventricular/fisiologia
20.
Alzheimers Dement (N Y) ; 7(1): e12130, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33521236

RESUMO

INTRODUCTION: This study assessed the hypothesis that circulating human amylin (amyloid-forming) cross-seeds with amyloid beta (Aß) in early Alzheimer's disease (AD). METHODS: Evidence of amylin-AD pathology interaction was tested in brains of 31 familial AD mutation carriers and 20 cognitively unaffected individuals, in cerebrospinal fluid (CSF) (98 diseased and 117 control samples) and in genetic databases. For functional testing, we genetically manipulated amylin secretion in APP/PS1 and non-APP/PS1 rats. RESULTS: Amylin-Aß cross-seeding was identified in AD brains. High CSF amylin levels were associated with decreased CSF Aß42 concentrations. AD risk and amylin gene are not correlated. Suppressed amylin secretion protected APP/PS1 rats against AD-associated effects. In contrast, hypersecretion or intravenous injection of human amylin in APP/PS1 rats exacerbated AD-like pathology through disruption of CSF-brain Aß exchange and amylin-Aß cross-seeding. DISCUSSION: These findings strengthened the hypothesis of circulating amylin-AD interaction and suggest that modulation of blood amylin levels may alter Aß-related pathology/symptoms.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA