Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Eur J Pharmacol ; 917: 174752, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-35026192

RESUMO

Gynaecological cancers continue to present a significant health burden upon the health of the global female population. This deficit is most prominent with ovarian cancer which possesses the lowest survival rate compared to all other cancers occurring within this anatomical region, with an annual UK-mortality of 7,300. The poor tolerability and selectively of the treatment options that are currently available is likely to have contributed to this high mortality rate thus, demonstrating the need for the development of enhanced therapeutic approaches. Aptamer technology would involve the engineering of specifically sequenced oligonucleotide chains, which bind to macromolecular targets with a high degree of affinity and selectively. Recent in-vitro studies conducted upon the clinical utility of this technique have supported its superiority in targeting individual therapeutic drug targets compared to various other targeting moieties currently within therapeutic use such as, monoclonal antibodies. For this reason, the employment of this technique is likely to be favourable in reducing the incidence of non-specific, chemotherapy-associated adverse effects. Kisspeptin is a naturally expressed polypeptide with an established role in the development of the reproductive system and other proposed roles in influencing the ability of ovarian cancer growths to exhibit the metastasis hallmark. This distinctive feature would indicate the potential for the manipulation of this pathway through the application of aptamer structures in developing a novel prophylactic strategy and improve the long-term outcome for ovarian cancer patients.


Assuntos
Kisspeptinas
2.
Int J Biol Macromol ; 174: 175-184, 2021 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-33516852

RESUMO

Protein disulfide isomerase (PDI) is an important molecular chaperone capable of facilitating protein folding in addition to catalyzing the formation of a disulfide bond. To better understand the distinct substrate-screening principles of Pichia pastoris PDI (Protein disulfide isomerase) and the protective role of PDI in amyloidogenic diseases, we investigated the expression abundance and intracellular retention levels of three archetypal amyloidogenic disulfide bond-free proteins (Aß42, α-synuclein (α-Syn) and SAA1) in P. pastoris GS115 strain without and with the overexpression of PpPDI (P. pastoris PDI). Intriguingly, amyloidogenic Aß42 and α-Syn were detected only as intracellular proteins whereas amyloidogenic SAA1 was detected both as intracellular and extracellular proteins when these proteins were expressed in the PpPDI-overexpressing GS115 strain. The binding between PpPDI and each of the three amyloidogenic proteins was investigated by molecular docking and simulations. Three different patterns of PpPDI-substrate complexes were observed, suggesting that multiple modes of binding might exist for the binding between PpPDI and its amyloidogenic protein substrates, and this could represent different specificities and affinities of PpPDI toward its substrates. Further analysis of the proteomics data and functional annotations indicated that PpPDI could eliminate the need for misfolded proteins to be partitioned in ER-associated compartments.


Assuntos
Isomerases de Dissulfetos de Proteínas/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Proteínas Amiloidogênicas/metabolismo , Cromatografia Líquida/métodos , Dissulfetos/química , Retículo Endoplasmático/metabolismo , Expressão Gênica/genética , Espectrometria de Massas/métodos , Chaperonas Moleculares/metabolismo , Simulação de Acoplamento Molecular , Pichia/enzimologia , Pichia/genética , Isomerases de Dissulfetos de Proteínas/fisiologia , Dobramento de Proteína , Processamento de Proteína Pós-Traducional/fisiologia , Proteômica/métodos , Saccharomycetales/enzimologia , Saccharomycetales/genética , Saccharomycetales/metabolismo , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
3.
Neural Regen Res ; 15(7): 1191-1198, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31960798

RESUMO

N-formyl peptide receptors (FPRs) were first identified upon phagocytic leukocytes, but more than four decades of research has unearthed a plethora of non-myeloid roles for this receptor family. FPRs are expressed within neuronal tissues and markedly in the central nervous system, where FPR interactions with endogenous ligands have been implicated in the pathophysiology of several neurodegenerative diseases including Alzheimer's disease and Parkinson's disease, as well as neurological cancers such as neuroblastoma. Whilst the homeostatic function of FPRs in the nervous system is currently undefined, a variety of novel physiological roles for this receptor family in the neuronal context have been posited in both human and animal settings. Rapid developments in recent years have implicated FPRs in the process of neurogenesis and neuronal differentiation which, upon greater characterisation, could represent a novel pharmacological target for neuronal regeneration therapies that may be used in the treatment of brain/spinal cord injury, stroke and neurodegeneration. This review aims to summarize the recent progress made to determine the physiological role of FPRs in a neuronal setting, and to put forward a case for FPRs as a novel pharmacological target for conditions of the nervous system, and for their potential to open the door to novel neuronal regeneration therapies.

4.
PLoS One ; 14(6): e0217815, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31170199

RESUMO

The N-formyl peptide receptors (FPRs) have been identified within neuronal tissues and may serve as yet undetermined functions within the nervous system. The FPRs have been implicated in the progression and invasiveness of neuroblastoma and other cancers. In this study the effects of the synthetic FPR agonist FPRa14, FPR antagonists and FPR knockdown using siRNA on mouse neuroblastoma neuro2a (N2a) cell differentiation plus toxicity were examined. The FPRa14 (1-10µM) was found to induce a significant dose-dependent differentiation response in mouse neuroblastoma N2a cells. Interestingly, three distinct differentiated morphologies were observed, with two non-archetypal forms observed at the higher FPRa14 concentrations. These three forms were also observed in the human neuroblastoma cell-lines IMR-32 and SH-SY5Y when exposed to 100µM FPRa14. In N2a cells combined knockdown of FPR1 and FPR2 using siRNA inhibited the differentiation response to FPRa14, suggesting involvement of both receptor subtypes. Pre-incubating N2a cultures with the FPR1 antagonists Boc-MLF and cyclosporin H significantly reduced FPRa14-induced differentiation to near baseline levels. Meanwhile, the FPR2 antagonist WRW4 had no significant effect on FPRa14-induced N2a differentiation. These results suggest that the N2a differentiation response observed has an FPR1-dependent component. Toxicity of FPRa14 was only observed at higher concentrations. All three antagonists used blocked FPRa14-induced toxicity, whilst only siRNA knockdown of FPR2 reduced toxicity. This suggests that the toxicity and differentiation involve different mechanisms. The demonstration of neuronal differentiation mediated via FPRs in this study represents a significant finding and suggests a role for FPRs in the CNS. This finding could potentially lead to novel therapies for a range of neurological conditions including neuroblastoma, Alzheimer's disease, Parkinson's disease and neuropathic pain. Furthermore, this could represent a potential avenue for neuronal regeneration therapies.


Assuntos
Diferenciação Celular , Forma Celular , Neuroblastoma/patologia , RNA Interferente Pequeno/metabolismo , Receptores de Formil Peptídeo/agonistas , Receptores de Formil Peptídeo/antagonistas & inibidores , Animais , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Humanos , Camundongos , Receptores de Formil Peptídeo/metabolismo , Fatores de Tempo
5.
Acta Histochem ; 116(6): 1022-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24835186

RESUMO

Histidine-tagged proteins are widely used in biochemical studies and frequently detected with antibodies specific for the histidine tag. Immunocytochemistry is widely used in studies with overexpressed proteins to determine cellular localization and in the case of histidine-tagged proteins can be carried out with anti-polyhistidine antibodies. Recent studies have suggested that polyhistidine sequences are present within a small number of human proteins and may direct expression to the nucleus and nuclear speckles compartments of the cell. In this study immunocytochemical staining of human SH-SY5Y neuroblastoma cell lines with the HIS-1 anti-polyhistidine monoclonal antibody were determined. Results showed that the HIS-1 anti-polyhistidine monoclonal antibody stained endogenous nuclear proteins in SH-SY5Y cells. The stained proteins were contained within the nuclear membrane, but were not directly linked to DNA. In a histidine-tagged catalase overexpressing cell line the HIS-1 anti-polyhistidine monoclonal antibody showed nuclear staining, whilst staining with the CAT-505 anti-catalase monoclonal antibody showed primarily cytoplasmic staining. These results suggest that anti-polyhistidine antibody staining shows significant cross-reactivity with endogenous nuclear proteins in SH-SY5Y neuroblastoma cells and may not be suitable for localization studies of histidine-tagged proteins. Immunocytochemical studies with anti-polyhistidine antibodies and localization of histidine-tagged proteins must be confirmed with protein specific antibodies or other methodology.


Assuntos
Anticorpos Monoclonais Murinos/química , Artefatos , Catalase/metabolismo , Linhagem Celular Tumoral , Técnica Indireta de Fluorescência para Anticorpo , Histidina/imunologia , Humanos , Proteínas Nucleares/metabolismo , Oligopeptídeos/imunologia , Proteínas Recombinantes de Fusão/metabolismo
6.
ACS Chem Neurosci ; 4(11): 1501-12, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-23968537

RESUMO

Alzheimer's disease, Familial British dementia, Familial Danish dementia, Type 2 diabetes mellitus, plus Creutzfeldt-Jakob disease are associated with amyloid fibril deposition and oxidative stress. The antioxidant enzyme catalase is a neuroprotective amyloid binding protein. Herein the effects of catalase overexpression in SH-SY5Y neuronal cells on the toxicity of amyloid-ß (Aß), amyloid-Bri (ABri), amyloid-Dan (ADan), amylin (IAPP), and prion protein (PrP) peptides were determined. Results showed catalase overexpression was neuroprotective against Aß, ABri, ADan, IAPP, and PrP peptides. The catalase inhibitor 3-amino-1,2,4-triazole (3-AT) and catalase-amyloid interaction inhibitor benzothiazole aniline tetra(ethylene glycol) (BTA-EG4) significantly enhanced neurotoxicity of amyloid peptides in catalase overexpressing neuronal cells. This suggests catalase neuroprotection involves breakdown of hydrogen peroxide (H2O2) plus a direct binding interaction between catalase and the Aß, ABri, ADan, IAPP, and PrP peptides. Kisspeptin 45-50 had additive neuroprotective actions against the Aß peptide in catalase overexpressing cells. The effects of 3-AT had an intracellular site of action, while catalase-amyloid interactions had an extracellular component. These results suggest that the 3-AT and BTA-EG4 compounds may be able to inhibit endogenous catalase mediated neuroprotection. Use of BTA-EG4, or compounds that inhibit catalase binding to amyloid peptides, as potential therapeutics for Neurodegenerative diseases may therefore result in unwanted effects.


Assuntos
Amitrol (Herbicida)/toxicidade , Peptídeos beta-Amiloides/antagonistas & inibidores , Compostos de Anilina/toxicidade , Benzotiazóis/toxicidade , Catalase/genética , Inibidores Enzimáticos/toxicidade , Fármacos Neuroprotetores/antagonistas & inibidores , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Amitrol (Herbicida)/química , Peptídeos beta-Amiloides/metabolismo , Compostos de Anilina/química , Benzotiazóis/química , Catalase/antagonistas & inibidores , Catalase/biossíntese , Linhagem Celular Tumoral , Síndrome de Creutzfeldt-Jakob/enzimologia , Síndrome de Creutzfeldt-Jakob/genética , Síndrome de Creutzfeldt-Jakob/patologia , Demência/enzimologia , Demência/genética , Demência/patologia , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Inibidores Enzimáticos/química , Humanos , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
7.
Micron ; 44: 246-53, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22854213

RESUMO

Full-length native rat amylin 1-37 has previously been widely shown to be unable to form fibrils and to lack the toxicity of the human amylin form leading to its use as a non-amyloidogenic control peptide. A recent study has suggested that rat amylin 1-37 forms amyloidogenic ß-sheet structures in the presence of the human amylin form and suggested that this property could promote toxicity. Using TEM analysis we show here fibril formation by synthetic rat amylin 1-37 and 8-37 peptides when the lyophilized HPLC purified peptides are initially dissolved in 20 mM Tris-HCl. Dissolution of synthetic rat amylin 1-37 and 8-37 peptides in H(2)O or phosphate buffered saline failed to produce fibrils. Addition of 20 mM Tris-HCl to synthetic rat amylin 1-37 and 8-37 peptides initially dissolved in H(2)O also failed to induce fibril formation. The rat amylin fibrils have a uniform structure and bind Congo red suggesting that they are amyloid fibrils. The rat amylin fibrils also bind catalase, which could be inhibited by Amyloid-ß 31-35 and a catalase amyloid-ß binding domain-like peptide (R9). The rat amylin 1-37 and 8-37 fibrils are toxic in both human pancreatic islet and neuronal cell culture systems. The toxicity of rat amylin fibrils can be inhibited by an amylin receptor antagonist (AC187) and a caspase inhibitor (zVAD-fmk) in a similar manner to previous observations for human amylin toxicity. Chemically induced rat amylin fibril formation of uniform structured fibrils provides a potentially novel anti-amyloid drug discovery tool.


Assuntos
Amiloide , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Amiloide/antagonistas & inibidores , Amiloide/biossíntese , Amiloide/metabolismo , Amiloide/toxicidade , Proteínas Amiloidogênicas , Animais , Catalase/metabolismo , Células Cultivadas , Humanos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Microscopia Eletrônica de Transmissão , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/farmacologia , Ratos
8.
J Neurodegener Dis ; 2013: 879710, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-26317001

RESUMO

The pons region of the Alzheimer's disease (AD) brain is one of the last to show amyloid-ß (Aß) deposits and has been suggested to contain neuroprotective compounds. Kisspeptin (KP) is a hormone that activates the hypothalamic-pituitary-gonadal axis and has been suggested to be neuroprotective against Aß toxicity. The localization of KP, plus the established endogenous neuroprotective compounds corticotropin releasing hormone (CRH) and catalase, in tissue sections from the pons region of a male AD subject has been determined in relation to Aß deposits. Results showed Aß deposits also stained with KP, CRH, and catalase antibodies. At high magnification the staining of deposits was either KP or catalase positive, and there was only a limited area of the deposits with KP-catalase colocalization. The CRH does not bind Aß, whilst both KP and catalase can bind Aß, suggesting that colocalization in Aß deposits is not restricted to compounds that directly bind Aß. The neuroprotective actions of KP, CRH, and catalase were confirmed in vitro, and fibrillar Aß preparations were shown to stimulate the release of KP in vitro. In conclusion, neuroprotective KP, CRH, and catalase all colocalize with Aß plaque-like deposits in the pons region from a male AD subject.

9.
ISRN Neurosci ; 2013: 253210, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24967306

RESUMO

Recent studies have suggested that the kisspeptin (KP) and kissorphin (KSO) peptides have neuroprotective actions against the Alzheimer's amyloid- ß (A ß ) peptide. Overexpression of the human KiSS-1 gene that codes for KP and KSO peptides in SH-SY5Y neurons has also been shown to inhibit A ß neurotoxicity. The in vivo actions of KP include activation of neuroendocrine and neurotransmitter systems. The present study used antagonists of KP, neuropeptide FF (NPFF), opioids, oxytocin, estrogen, adrenergic, cholinergic, dopaminergic, serotonergic, and γ -aminobutyric acid (GABA) receptors plus inhibitors of catalase, cyclooxygenase, nitric oxide synthase, and the mitogen activated protein kinase cascade to characterize the KiSS-1 gene overexpression neuroprotection against A ß cell model. The results showed that KiSS-1 overexpression is neuroprotective against A ß and the action appears to involve the KP or KSO peptide products of KiSS-1 processing. The mechanism of neuroprotection does not involve the activation of the KP or NPFF receptors. Opioids play a role in the toxicity of A ß in the KiSS-1 overexpression system and opioid antagonists naloxone or naltrexone inhibited A ß toxicity. The mechanism of KiSS-1 overexpression induced protection against A ß appears to have an oxytocin plus a cyclooxygenase dependent component, with the oxytocin antagonist atosiban and the cyclooxygenase inhibitor SC-560 both enhancing the toxicity of A ß .

10.
Subcell Biochem ; 65: 3-25, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23224997

RESUMO

In this chapter we provided the overall background to the subject of protein aggregation and fibrillogenesis in amyloidogenesis, with introduction and brief discussion of the various topics that are included with the coming chapters. The division of the book into basic science and clinical science sections enables correlation of the topics to be made. The many proteins and peptides that have currently been found to undergo fibrillogenesis are tabulated. A broad technical survey is made, to indicate the vast array of techniques currently available to study aspects of protein oligomerization, aggregation and fibrillogenesis. These are split into three groups and tabulated, as the microscopical techniques, the analytical and biophysical methods, and the biochemical and cellular techniques. A few techniques are discussed, but in most cases only a link to relevant recent literature is provided.


Assuntos
Amiloide/química , Peptídeos/química , Multimerização Proteica , Amiloide/ultraestrutura , Animais , Bioquímica/métodos , Biofísica/métodos , Humanos , Microscopia/métodos
11.
ACS Chem Neurosci ; 3(9): 706-19, 2012 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-23019497

RESUMO

Alzheimer's disease (AD) onset is associated with changes in hypothalamic-pituitary-gonadal (HPG) function. The 54 amino acid kisspeptin (KP) peptide regulates the HPG axis and alters antioxidant enzyme expression. The Alzheimer's amyloid-ß (Aß) is neurotoxic, and this action can be prevented by the antioxidant enzyme catalase. Here, we examined the effects of KP peptides on the neurotoxicity of Aß, prion protein (PrP), and amylin (IAPP) peptides. The Aß, PrP, and IAPP peptides stimulated the release of KP and KP 45-54. The KP peptides inhibited the neurotoxicity of Aß, PrP, and IAPP peptides, via an action that could not be blocked by kisspeptin-receptor (GPR-54) or neuropeptide FF (NPFF) receptor antagonists. Knockdown of KiSS-1 gene, which encodes the KP peptides, in human neuronal SH-SY5Y cells with siRNA enhanced the toxicity of amyloid peptides, while KiSS-1 overexpression was neuroprotective. A comparison of the catalase and KP sequences identified a similarity between KP residues 42-51 and the region of catalase that binds Aß. The KP peptides containing residues 45-50 bound Aß, PrP, and IAPP, inhibited Congo red binding, and were neuroprotective. These results suggest that KP peptides are neuroprotective against Aß, IAPP, and PrP peptides via a receptor independent action involving direct binding to the amyloid peptides.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Kisspeptinas/farmacologia , Neurotoxinas/antagonistas & inibidores , Neurotoxinas/toxicidade , Peptídeos beta-Amiloides/metabolismo , Animais , Western Blotting , Catalase/antagonistas & inibidores , Morte Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Corantes , Vermelho Congo , Humanos , Técnicas Imunoenzimáticas , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Cinética , Kisspeptinas/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fosforilação , Príons/antagonistas & inibidores , Príons/toxicidade , Ligação Proteica , RNA Interferente Pequeno/genética , Ratos
12.
J Amino Acids ; 2012: 691463, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22848794

RESUMO

The primary products of the metastasis-suppressor KiSS-1 gene are the kisspeptin (KP) peptides that stimulate gonadotrophin-releasing-hormone (GnRH) release via GPR-54 receptor activation. Recent studies have suggested that the KP-10 peptide also activates neuropeptide FF (NPFF) receptors. The aim of the current study was to determine the activities of the KiSS-1 derivative kissorphin (KSO), which contains the first six amino acids of the KP-10 peptide, is C-terminally amidated, and shares amino acid similarities with the biologically active NPFF 3-8 sequence. The KSO peptide inhibited forskolin-stimulated cyclic adenosine monophosphate (cAMP) production in ND7/23 neuroblastoma cells via an action that could be inhibited by the NPFF receptor antagonist RF9. Release of GnRH by LA-N-1 neuroblastoma cells was not altered by the KSO peptide. In ND7/23 neuroblastoma cells, the KSO peptide was able to reduce forskolin neuroprotection against H(2)O(2) toxicity. The KSO peptide was also able to prevent prostaglandin E2-induced apoptosis in rat cortical neurons. The NPFF receptor antagonist RF9 could inhibit these actions of the KSO peptide in oxidative stress and apoptosis models. In conclusion, the kissorphin peptide, comprising the amino acid sequence Tyr-Asn-Trp-Asn-Ser-Phe-NH(2), has NPFF-like biological activity without showing any GnRH releasing activity and inhibits forskolin-activated cAMP release.

13.
ScientificWorldJournal ; 10: 879-93, 2010 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-20495767

RESUMO

The diabetes-associated human islet amyloid polypeptide (IAPP) is a 37-amino-acid peptide that forms fibrils in vitro and in vivo. Human IAPP fibrils are toxic in a similar manner to Alzheimer's amyloid-beta (A-beta) and prion protein (PrP) fibrils. Previous studies have shown that catalase binds to A-beta fibrils and appears to recognize a region containing the Gly-Ala-Ile-Ile sequence that is similar to the Gly-Ala-Ile-Leu sequence found in human IAPP residues 24-27. This study presents a transmission electron microscopy (TEM)-based analysis of fibril formation and the binding of human erythrocyte catalase to IAPP fibrils. The results show that human IAPP 1-37, 8-37, and 20-29 peptides form fibrils with diverse and polymorphic structures. All three forms of IAPP bound catalase, and complexes of IAPP 1-37 or 8-37 with catalase were identified by immunoassay. The binding of biotinylated IAPP to catalase was high affinity with a KD of 0.77 nM, and could be inhibited by either human or rat IAPP 1-37 and 8-37 forms. Fibrils formed by the PrP 118-135 peptide with a Gly-Ala-Val-Val sequence also bound catalase. These results suggest that catalase recognizes a Gly-Ala-Ile-Leu-like sequence in amyloid fibril-forming peptides. For IAPP 1-37 and 8-37, the catalase binding was primarily directed towards fibrillar rather than ribbon-like structures, suggesting differences in the accessibility of the human IAPP 24-27 Gly-Ala-Ile-Leu region. This suggests that catalase may be able to discriminate between different structural forms of IAPP fibrils. The ability of catalase to bind IAPP, A-beta, and PrP fibrils demonstrates the presence of similar accessible structural motifs that may be targets for antiamyloid therapeutic development.


Assuntos
Amiloide/metabolismo , Catalase/metabolismo , Amiloide/ultraestrutura , Animais , Catalase/sangue , Ensaio de Imunoadsorção Enzimática , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Microscopia Eletrônica de Transmissão , Ligação Proteica , Ratos
14.
Subcell Biochem ; 51: 47-75, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20213540

RESUMO

The complex association of cholesterol metabolism and Alzheimer's disease is presented in depth, including the possible benefits to be gained from cholesterol-lowering statin therapy. Then follows a survey of the role of neuronal membrane cholesterol in Abeta pore formation and Abeta fibrillogenesis, together with the link with membrane raft domains and gangliosides. The contribution of structural studies to Abeta fibrillogenesis, using TEM and AFM, is given some emphasis. The role of apolipoprotein E and its isoforms, in particular ApoE4, in cholesterol and Abeta binding is presented, in relation to genetic risk factors for Alzheimer's disease. Increasing evidence suggests that cholesterol oxidation products are of importance in generation of Alzheimer's disease, possibly induced by Abeta-produced hydrogen peroxide. The body of evidence for a link between cholesterol in atherosclerosis and Alzheimer's disease is increasing, along with an associated inflammatory response. The possible role of cholesterol in tau fibrillization, tauopathies and in some other non-Abeta amyloidogenic disorders is surveyed.


Assuntos
Doença de Alzheimer/fisiopatologia , Colesterol/metabolismo , Doença de Alzheimer/complicações , Doença de Alzheimer/tratamento farmacológico , Amiloide/biossíntese , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Apolipoproteínas E/metabolismo , Aterosclerose , Encéfalo/metabolismo , Gangliosídeo G(M1)/fisiologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Masculino , Neurônios/metabolismo , Estresse Oxidativo/fisiologia , Tauopatias/fisiopatologia , Proteínas tau/metabolismo
15.
Micron ; 40(8): 800-10, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19683452

RESUMO

The Alzheimer's amyloid-beta (Abeta) peptide exists as a number of naturally occurring forms due to differential proteolytic processing of its precursor molecule. Many of the Abeta peptides of different lengths form fibrils in vitro, which often show polymorphisms in the fibril structure. This study presents a TEM based analysis of fibril formation by eighteen different Abeta peptides ranging in length from 5 to 43 amino acids. Spectrophotometric analysis of Congo red binding to the fibrillar material has been assessed and the binding of human erythrocyte catalase (HEC) to Abeta fibrils has also been investigated by TEM. The results show that a diverse range of Abeta peptides form fibrils and also bind Congo red. The ability of both Abeta 1-28 and Abeta 29-40 to form fibrils indicates that there are at least two fibril-forming domains within the full-length Abeta 1-40 sequence, the ability of many Abeta peptides to form Congo red-binding aggregates suggests that there may be up to 4 possible aggregation promoting domains. The binding of HEC was limited to Abeta forms containing residues 29-32. The differing capacities of fibrillar and ribbon-like structures may reflect the accessibility of the 29-32 region and suggest that HEC may be able to discriminate between different forms of Abeta fibrils.


Assuntos
Amiloide/ultraestrutura , Catalase/metabolismo , Eritrócitos/enzimologia , Vermelho Congo/metabolismo , Humanos , Microscopia Eletrônica de Transmissão , Ligação Proteica , Espectrofotometria/métodos , Coloração e Rotulagem/métodos
16.
Subcell Biochem ; 38: 381-402, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15709490

RESUMO

Phosphorylated Amyloid-beta (Abeta) was identified in Alzheimer's disease (AD) brain. Using an anti-sense peptide approach the human cyclin-dependent kinase-1 (CDK-1) was identified as being responsible for Abeta phosphorylation. The phosphorylated Abeta peptide showed increased neurotoxicity and reduced ability to form Congo red-positive fibrils. Mutation of the serine 26 residue and inhibition of Abeta phosphorylation by the CDK-1 inhibitor olomoucine prevented Abeta toxicity, suggesting that the phosphorylated Abeta peptide represents a toxic intermediate. Cannabinoids prevented phosphorylated Abeta toxicity. The results from this study suggest that Abeta phosphorylation could play a role in AD pathology and represent a novel therapeutic target.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Fosfoproteínas/toxicidade , Peptídeos beta-Amiloides/toxicidade , Animais , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/uso terapêutico , Humanos , Cinetina , Degeneração Neural/patologia , Fosfoproteínas/antagonistas & inibidores , Fosforilação , Purinas/uso terapêutico
17.
Drugs Aging ; 21(2): 81-100, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14960126

RESUMO

Hydrogen peroxide (H(2)O(2)) is a stable, uncharged and freely diffusable reactive oxygen species (ROS) and second messenger. The generation of H(2)O(2) in the brain is relatively high because of the high oxygen consumption in the tissue. Alzheimer's disease is a neurodegenerative disorder characterised by the appearance of amyloid-beta (Abeta)-containing plaques and hyperphosphorylated tau-containing neurofibrillary tangles. The pathology of Alzheimer's disease is also associated with oxidative stress and H(2)O(2) is implicated in this and the neurotoxicity of the Abeta peptide. The ability for Abeta to generate H(2)O(2), and interactions of H(2)O(2) with iron and copper to generate highly toxic ROS, may provide a mechanism for the oxidative stress associated with Alzheimer's disease. The role of heavy metals in Alzheimer's disease pathology and the toxicity of the H(2)O(2) molecule may be closely linked. Drugs that prevent oxidative stress include antioxidants, modifiers of the enzymes involved in ROS generation and metabolism, metal chelating agents and agents that can remove the stimulus for ROS generation. In Alzheimer's disease the H(2)O(2) molecule must be considered a therapeutic target for treatment of the oxidative stress associated with the disease. The actions of H(2)O(2) include modifications of proteins, lipids and DNA, all of which are effects seen in the Alzheimer's disease brain and may contribute to the loss of synaptic function characteristic of the disease. The effectiveness of drugs to target this component of the disease pathology remains to be determined; however, metal chelators may provide an effective route and have the added bonus in the case of clioquinol of potentially reducing the Abeta load. Future research and development of agents that specifically target the H(2)O(2) molecule or enzymes involved in its metabolism may provide the future route to Alzheimer's disease therapy.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/fisiologia , Antioxidantes/uso terapêutico , Encéfalo , Quelantes/uso terapêutico , Peróxido de Hidrogênio/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/etiologia , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Encéfalo/fisiologia , Humanos , Peróxido de Hidrogênio/antagonistas & inibidores
18.
Neurosci Lett ; 332(2): 127-30, 2002 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-12384227

RESUMO

Cannabinoid receptor agonists including anandamide and noladin either have recently been suggested to exhibit neuroprotective properties. The amyloid-beta (Abeta) peptide is thought to be responsible for the neurodegenerative changes associated with Alzheimer's disease pathology. This study characterizes the effects of anandamide and noladin ether on the neurotoxicity of Abeta in differentiated human teratocarcinoma cell line, Ntera 2/cl-D1 neurons. Anandamide and noladin ether, at nanomolar concentrations, showed concentration dependent inhibition of Abeta toxicity. A CB(1) cannabinoid receptor antagonist, AM251, prevented the protective effects of anandamide and noladin ether. The mitogen activated protein kinase (MAPK) pathway inhibitor PD98059 also prevented the protective effects of cannabinoids and corticotrophin-releasing hormone. These results suggest that activation of the MAPK pathway by either cannabinoids or corticotrophin-releasing hormone could be used to prevent Abeta peptide induced neurodegeneration.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Ácidos Araquidônicos/farmacologia , Canabinoides/farmacologia , Glicerídeos/farmacologia , Doenças do Sistema Nervoso/induzido quimicamente , Doenças do Sistema Nervoso/prevenção & controle , Fármacos Neuroprotetores , Neurotoxinas/antagonistas & inibidores , Neurotoxinas/toxicidade , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/toxicidade , Hormônio Liberador da Corticotropina/farmacologia , Endocanabinoides , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Humanos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Neurônios/patologia , Oxirredução , Fragmentos de Peptídeos/farmacologia , Alcamidas Poli-Insaturadas , Receptores de Canabinoides , Receptores de Droga/antagonistas & inibidores
19.
Neurosci Lett ; 322(2): 131-3, 2002 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-11958860

RESUMO

The pathological features of Alzheimer's disease include deposition of amyloid-beta (Abeta) containing plaques and increases in the expression of cyclin-dependent kinase (CDK) enzymes. Chemical inhibition of CDKs prevents the neurotoxicity of the Abeta peptide. The activity of these kinases requires the binding of a cyclin component to the catalytic enzyme component. This study characterizes direct interactions between Abeta and cyclin B1. Abeta fragments containing the cytotoxic 31-35 region could inhibit biotinylated Abeta binding to cyclin B1. The same cytotoxic Abeta fragments all increased CDK-1 phosphorylation of known substrates in a cell free system. The CDK-1 inhibitor olomoucine prevented the cytotoxicity of Abeta 31-35 containing peptides in differentiated human teratocarcinoma cell line, Ntera 2/cl-D1 (NT-2) neurons. These direct interactions between cyclin B1 and Abeta provide potential mechanisms for the cytotoxicity of the Abeta peptide.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Proteína Quinase CDC2/metabolismo , Ciclina B/metabolismo , Fragmentos de Peptídeos/metabolismo , Sequência de Aminoácidos , Ciclina B1 , Humanos , Dados de Sequência Molecular , Neurônios/metabolismo , Ligação Proteica , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA