Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 13: 821091, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35299958

RESUMO

Toxic misfolding of proinsulin variants in ß-cells defines a monogenic diabetes syndrome, designated mutant INS-gene induced diabetes of the young (MIDY). In our first study (previous article in this issue), we described a one-disulfide peptide model of a proinsulin folding intermediate and its use to study such variants. The mutations (LeuB15→Pro, LeuA16→Pro, and PheB24→Ser) probe residues conserved among vertebrate insulins. In this companion study, we describe 1H and 1H-13C NMR studies of the peptides; key NMR resonance assignments were verified by synthetic 13C-labeling. Parent spectra retain nativelike features in the neighborhood of the single disulfide bridge (cystine B19-A20), including secondary NMR chemical shifts and nonlocal nuclear Overhauser effects. This partial fold engages wild-type side chains LeuB15, LeuA16 and PheB24 at the nexus of nativelike α-helices α1 and α3 (as defined in native proinsulin) and flanking ß-strand (residues B24-B26). The variant peptides exhibit successive structural perturbations in order: parent (most organized) > SerB24 >> ProA16 > ProB15 (least organized). The same order pertains to (a) overall α-helix content as probed by circular dichroism, (b) synthetic yields of corresponding three-disulfide insulin analogs, and (c) ER stress induced in cell culture by corresponding mutant proinsulins. These findings suggest that this and related peptide models will provide a general platform for classification of MIDY mutations based on molecular mechanisms by which nascent disulfide pairing is impaired. We propose that the syndrome's variable phenotypic spectrum-onsets ranging from the neonatal period to later in childhood or adolescence-reflects structural features of respective folding intermediates.


Assuntos
Diabetes Mellitus , Proinsulina , Adolescente , Diabetes Mellitus/genética , Dissulfetos/química , Humanos , Recém-Nascido , Insulina/química , Proinsulina/química , Proinsulina/genética , Dobramento de Proteína
2.
Front Endocrinol (Lausanne) ; 13: 821069, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35299972

RESUMO

The mutant proinsulin syndrome is a monogenic cause of diabetes mellitus due to toxic misfolding of insulin's biosynthetic precursor. Also designated mutant INS-gene induced diabetes of the young (MIDY), this syndrome defines molecular determinants of foldability in the endoplasmic reticulum (ER) of ß-cells. Here, we describe a peptide model of a key proinsulin folding intermediate and variants containing representative clinical mutations; the latter perturb invariant core sites in native proinsulin (LeuB15→Pro, LeuA16→Pro, and PheB24→Ser). The studies exploited a 49-residue single-chain synthetic precursor (designated DesDi), previously shown to optimize in vitro efficiency of disulfide pairing. Parent and variant peptides contain a single disulfide bridge (cystine B19-A20) to provide a model of proinsulin's first oxidative folding intermediate. The peptides were characterized by circular dichroism and redox stability in relation to effects of the mutations on (a) in vitro foldability of the corresponding insulin analogs and (b) ER stress induced in cell culture on expression of the corresponding variant proinsulins. Striking correlations were observed between peptide biophysical properties, degree of ER stress and age of diabetes onset (neonatal or adolescent). Our findings suggest that age of onset reflects the extent to which nascent structure is destabilized in proinsulin's putative folding nucleus. We envisage that such peptide models will enable high-resolution structural studies of key folding determinants and in turn permit molecular dissection of phenotype-genotype relationships in this monogenic diabetes syndrome. Our companion study (next article in this issue) employs two-dimensional heteronuclear NMR spectroscopy to define site-specific perturbations in the variant peptides.


Assuntos
Diabetes Mellitus , Proinsulina , Adolescente , Diabetes Mellitus/metabolismo , Dissulfetos/química , Dissulfetos/metabolismo , Humanos , Insulina/metabolismo , Peptídeos , Proinsulina/química , Proinsulina/genética , Proinsulina/metabolismo , Dobramento de Proteína
3.
Diabetes ; 70(12): 2947-2956, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34649926

RESUMO

Human insulin (INS) gene diverged from the ancestral genes of invertebrate and mammalian species millions of years ago. We previously found that mouse insulin gene (Ins2) isoforms are expressed in brain choroid plexus (ChP) epithelium cells, where insulin secretion is regulated by serotonin and not by glucose. We further compared human INS isoform expression in postmortem ChP and islets of Langerhans. We uncovered novel INS upstream open reading frame isoforms and their protein products. In addition, we found a novel alternatively spliced isoform that translates to a 74-amino acid (AA) proinsulin containing a shorter 19-AA C-peptide sequence, herein designated Cα-peptide. The middle portion of the conventional C-peptide contains ß-sheet (GQVEL) and hairpin (GGGPG) motifs that are not present in Cα-peptide. Islet amyloid polypeptide (IAPP) is not expressed in ChP, and its amyloid formation was inhibited in vitro more efficiently by Cα-peptide than by C-peptide. Of clinical relevance, the ratio of the 74-AA proinsulin to proconvertase-processed Cα-peptide was significantly increased in islets from type 2 diabetes mellitus autopsy donors. Intriguingly, 100 years after the discovery of insulin, we found that INS isoforms are present in ChP from insulin-deficient autopsy donors.


Assuntos
Peptídeo C/metabolismo , Plexo Corióideo/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Adulto , Sequência de Aminoácidos , Amiloide/análise , Amiloide/química , Amiloide/metabolismo , Animais , Autopsia , Peptídeo C/análise , Peptídeo C/química , Plexo Corióideo/química , Plexo Corióideo/patologia , Humanos , Insulina/análise , Insulina/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/análise , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/química , Ilhotas Pancreáticas/patologia , Camundongos , Proinsulina/análise , Proinsulina/química , Proinsulina/metabolismo , Isoformas de Proteínas/análise , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo
4.
Clin Chem ; 67(6): 854-862, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34051096

RESUMO

BACKGROUND: Determination of C-peptide is important in the investigation of unexplained hyperinsulinemic hypoglycemia because a high C-peptide concentration usually indicates endogenous insulin hypersecretion. Insulin autoimmune syndrome (IAS) denotes hyperinsulinemic hypoglycemia due to insulin-binding antibodies that prolong insulin half-life. C-peptide clearance is considered to be unaffected, and although a marked C-peptide immunoreactivity in hypoglycemic samples has been reported, it has been suspected to be artifactual. High-resolution mass spectrometry enables examination of the basis of C-peptide-immunoreactivity in IAS. METHODS: Precipitation of plasma with polyethylene glycol was followed by C-peptide immunoassay. Plasma peptides extracted by solvent precipitation were characterized by nano-LC-MS/MS and analyzed using an untargeted data-dependent method. Peptides related to proinsulin, in amino acid sequence, were identified using proprietary bioinformatics software and confirmed by repeat LC-MS/MS analysis. Gel filtration chromatography coupled to LC-MS/MS was used to identify proinsulin-related peptides present in IAS immunocomplexes. Results were compared with those from C-peptide immunoassay. RESULTS: Polyethylene glycol precipitation of IAS plasma, but not control plasma, depleted C-peptide immunoreactivity consistent with immunoglobulin-bound C-peptide immunoreactivity. LC-MS/MS detected proinsulin and des 31,32 proinsulin at higher abundance in IAS plasma compared with control plasma. Analysis by gel filtration chromatography coupled to LC-MS/MS demonstrated proinsulin and des 31,32 proinsulin, but no C-peptide, in plasma immunocomplexes. CONCLUSIONS: Antibody binding can enrich proinsulin and des 31,32 proinsulin in IAS immunocomplexes. Proinsulin cross-reactivity in some C-peptide immunoassays can lead to artifactually increased C-peptide results.


Assuntos
Doenças Autoimunes , Hiperinsulinismo , Hipoglicemia , Anticorpos Anti-Insulina/química , Insulina/química , Peptídeos/química , Peptídeo C/química , Cromatografia Líquida , Humanos , Insulina/metabolismo , Peso Molecular , Polietilenoglicóis/química , Proinsulina/química , Espectrometria de Massas em Tandem
5.
FEBS J ; 287(20): 4440-4457, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32077246

RESUMO

Pro-islet amyloid polypeptide (proIAPP) is the prohormone precursor molecule to IAPP, also known as amylin. IAPP is a calcitonin family peptide hormone that is cosecreted with insulin, and largely responsible for hunger satiation and metabolic homeostasis. Amyloid plaques containing mixtures of mature IAPP and misprocessed proIAPP deposit on, and destroy pancreatic ß-cell membranes, and they are recognized as a clinical hallmark of type 2 diabetes mellitus. In order to better understand the interaction with cellular membranes, we solved the solution NMR structure of proIAPP bound to dodecylphosphocholine micelles at pH 4.5. We show that proIAPP is a dynamic molecule with four α-helices. The first two helices are contained within the mature IAPP sequence, while the second two helices are part of the C-terminal prohormone segment (Cpro). We mapped the membrane topology of the amphipathic helices by paramagnetic relaxation enhancement, and we used CD and diffusion-ordered spectroscopy to identify environmental factors that impact proIAPP membrane affinity. We discuss how our structural results relate to prohormone processing based on the varied pH environments and lipid compositions of organelle membranes within the regulated secretory pathway, and the likelihood of Cpro survival for cosecretion with IAPP. DATABASE: The assigned resonances have been deposited in the Biological Magnetic Resonance Bank (BMRB) with accession numbers 50007 and 50019 for proIAPP and Cpro, respectively. The lowest energy structures have been deposited in the Protein Data Bank (PDB) with access codes 6UCJ and 6UCK.


Assuntos
Amiloide/química , Proinsulina/química , Humanos , Concentração de Íons de Hidrogênio , Micelas , Simulação de Dinâmica Molecular , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica
6.
Int J Biol Macromol ; 139: 647-653, 2019 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-31356950

RESUMO

The present study deals with mass spectrometric investigation to characterize the nature of proinsulin in inclusion bodies. Various derivatives of human proinsulin were cloned, expressed in E. coli and inclusion bodies prepared under weak acidic conditions (pH 6.5), which protected the native thiols. Non-reductive PAGE showed that proinsulin migrated as monomer (approximately 10 kDa). MALDI-MS protocol was developed for the direct analysis of proinsulin derivatives in inclusion bodies. It was found that the masses of the derivatives corresponded to polypeptides containing six cysteines in reduced form. Iodoacetamide or iodoacetic acid treatment of proinsulin inclusion bodies, in suspension under non-reducing conditions and without any chaotropic agents, showed six alkylations, suggesting that these cytoplasmic aggregates were assembled from reduced monomers, with their -SH groups pointing towards hydrophilic surface. The MALDI analysis of inclusion bodies was extended to a proinsulin derivatives labelled with 13C and 15N giving an excellent agreement between experimental and theoretical masses. These mass spectrometric studies also provide early information about post-translational modification as evident in one of the derivatives MTRR-pi showing N-terminal cleavage of methionine. This shows the potential value of the protocol for the accurate analysis of polypeptides, expressed as inclusion bodies, prior to undertaking further purification.


Assuntos
Corpos de Inclusão/química , Proinsulina/química , Proteínas Recombinantes/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Isótopos de Carbono/química , Cisteína/química , Citoplasma/metabolismo , Escherichia coli , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Iodoacetamida/química , Metionina/química , Isótopos de Nitrogênio/química , Oxirredução , Peptídeos/química , Domínios Proteicos , Processamento de Proteína Pós-Traducional , Compostos de Sulfidrila/química
7.
Elife ; 82019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31184302

RESUMO

Biosynthesis of insulin - critical to metabolic homeostasis - begins with folding of the proinsulin precursor, including formation of three evolutionarily conserved intramolecular disulfide bonds. Remarkably, normal pancreatic islets contain a subset of proinsulin molecules bearing at least one free cysteine thiol. In human (or rodent) islets with a perturbed endoplasmic reticulum folding environment, non-native proinsulin enters intermolecular disulfide-linked complexes. In genetically obese mice with otherwise wild-type islets, disulfide-linked complexes of proinsulin are more abundant, and leptin receptor-deficient mice, the further increase of such complexes tracks with the onset of islet insulin deficiency and diabetes. Proinsulin-Cys(B19) and Cys(A20) are necessary and sufficient for the formation of proinsulin disulfide-linked complexes; indeed, proinsulin Cys(B19)-Cys(B19) covalent homodimers resist reductive dissociation, highlighting a structural basis for aberrant proinsulin complex formation. We conclude that increased proinsulin misfolding via disulfide-linked complexes is an early event associated with prediabetes that worsens with ß-cell dysfunction in type two diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Proinsulina/química , Dobramento de Proteína , Animais , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Progressão da Doença , Dissulfetos/química , Dissulfetos/metabolismo , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Humanos , Ilhotas Pancreáticas/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Proinsulina/genética , Proinsulina/metabolismo , Receptores para Leptina/deficiência , Receptores para Leptina/genética
8.
Elife ; 72018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30412052

RESUMO

Insulin gene mutations are a leading cause of neonatal diabetes. They can lead to proinsulin misfolding and its retention in endoplasmic reticulum (ER). This results in increased ER-stress suggested to trigger beta-cell apoptosis. In humans, the mechanisms underlying beta-cell failure remain unclear. Here we show that misfolded proinsulin impairs developing beta-cell proliferation without increasing apoptosis. We generated induced pluripotent stem cells (iPSCs) from people carrying insulin (INS) mutations, engineered isogenic CRISPR-Cas9 mutation-corrected lines and differentiated them to beta-like cells. Single-cell RNA-sequencing analysis showed increased ER-stress and reduced proliferation in INS-mutant beta-like cells compared with corrected controls. Upon transplantation into mice, INS-mutant grafts presented reduced insulin secretion and aggravated ER-stress. Cell size, mTORC1 signaling, and respiratory chain subunits expression were all reduced in INS-mutant beta-like cells, yet apoptosis was not increased at any stage. Our results demonstrate that neonatal diabetes-associated INS-mutations lead to defective beta-cell mass expansion, contributing to diabetes development.


Assuntos
Diabetes Mellitus/genética , Estresse do Retículo Endoplasmático/genética , Células-Tronco Pluripotentes Induzidas/química , Proinsulina/genética , Animais , Apoptose/genética , Sistemas CRISPR-Cas/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Diabetes Mellitus/patologia , Retículo Endoplasmático/genética , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Recém-Nascido , Células Secretoras de Insulina/química , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Mutação , Proinsulina/química , Dobramento de Proteína , Análise de Sequência de RNA , Transdução de Sinais , Análise de Célula Única
9.
Diabetes Obes Metab ; 20 Suppl 2: 28-50, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30230185

RESUMO

Insulin synthesis in pancreatic ß-cells is initiated as preproinsulin. Prevailing glucose concentrations, which oscillate pre- and postprandially, exert major dynamic variation in preproinsulin biosynthesis. Accompanying upregulated translation of the insulin precursor includes elements of the endoplasmic reticulum (ER) translocation apparatus linked to successful orientation of the signal peptide, translocation and signal peptide cleavage of preproinsulin-all of which are necessary to initiate the pathway of proper proinsulin folding. Evolutionary pressures on the primary structure of proinsulin itself have preserved the efficiency of folding ("foldability"), and remarkably, these evolutionary pressures are distinct from those protecting the ultimate biological activity of insulin. Proinsulin foldability is manifest in the ER, in which the local environment is designed to assist in the overall load of proinsulin folding and to favour its disulphide bond formation (while limiting misfolding), all of which is closely tuned to ER stress response pathways that have complex (beneficial, as well as potentially damaging) effects on pancreatic ß-cells. Proinsulin misfolding may occur as a consequence of exuberant proinsulin biosynthetic load in the ER, proinsulin coding sequence mutations, or genetic predispositions that lead to an altered ER folding environment. Proinsulin misfolding is a phenotype that is very much linked to deficient insulin production and diabetes, as is seen in a variety of contexts: rodent models bearing proinsulin-misfolding mutants, human patients with Mutant INS-gene-induced Diabetes of Youth (MIDY), animal models and human patients bearing mutations in critical ER resident proteins, and, quite possibly, in more common variety type 2 diabetes.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/biossíntese , Precursores de Proteínas/biossíntese , Animais , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Humanos , Insulina/química , Camundongos , Mutação/genética , Proinsulina/biossíntese , Proinsulina/química , Proinsulina/genética , Dobramento de Proteína , Precursores de Proteínas/química , Sistemas de Translocação de Proteínas/metabolismo
10.
J Cell Biol ; 217(4): 1287-1301, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29507125

RESUMO

In mammalian pancreatic ß cells, the IRE1α-XBP1 pathway is constitutively and highly activated under physiological conditions. To elucidate the precise role of this pathway, we constructed ß cell-specific Ire1α conditional knockout (CKO) mice and established insulinoma cell lines in which Ire1α was deleted using the Cre-loxP system. Ire1α CKO mice showed the typical diabetic phenotype including impaired glycemic control and defects in insulin biosynthesis postnatally at 4-20 weeks. Ire1α deletion in pancreatic ß cells in mice and insulinoma cells resulted in decreased insulin secretion, decreased insulin and proinsulin contents in cells, and decreased oxidative folding of proinsulin along with decreased expression of five protein disulfide isomerases (PDIs): PDI, PDIR, P5, ERp44, and ERp46. Reconstitution of the IRE1α-XBP1 pathway restored the proinsulin and insulin contents, insulin secretion, and expression of the five PDIs, indicating that IRE1α functions as a key regulator of the induction of catalysts for the oxidative folding of proinsulin in pancreatic ß cells.


Assuntos
Endorribonucleases/metabolismo , Células Secretoras de Insulina/enzimologia , Insulina/metabolismo , Proinsulina/metabolismo , Dobramento de Proteína , Proteínas Serina-Treonina Quinases/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Fator 6 Ativador da Transcrição/metabolismo , Animais , Sítios de Ligação , Glicemia/metabolismo , Linhagem Celular Tumoral , Diabetes Mellitus/sangue , Diabetes Mellitus/enzimologia , Diabetes Mellitus/genética , Endorribonucleases/deficiência , Endorribonucleases/genética , Insulina/genética , Insulinoma/enzimologia , Insulinoma/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Knockout , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Oxirredução , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Fosforilação , Proinsulina/química , Proinsulina/genética , Regiões Promotoras Genéticas , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Proteína 1 de Ligação a X-Box/genética , eIF-2 Quinase/metabolismo
11.
Protein Expr Purif ; 146: 91-96, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29432812

RESUMO

This paper describes a new pH-responsive peptide tag that adds a protein reversible precipitation and redissolution character. This peptide tag is a part of a cell surface protein B (CspB) derived from Corynebacterium glutamicum. Proinsulin that genetically fused with a peptide of N-terminal 6, 17, 50, or 250 amino acid residues of CspB showed that the reversible precipitation and redissolution depended on the pH. The transition occurred within a physiological and narrow pH range. A CspB50 tag comprising 50 amino acid residues of N-terminal CspB was further evaluated as a representative using other pharmaceutical proteins. Below pH 6.8, almost all CspB50-Teriparatide fusion formed an aggregated state. Subsequent addition of alkali turned the cloudy protein solution transparent above pH 7.3, in which almost all the CspB50-Teriparatide fusion redissolved. The CspB50-Bivalirudin fusion showed a similar behavior with slightly different pH range. This tag is offering a new protein purification method based on liquid-solid separation which does not require an affinity ligand. This sharp response around neutral pH is useful as a pH-responsive tag for the purification of unstable proteins at a non-physiological pH.


Assuntos
Proteínas de Bactérias/química , Corynebacterium glutamicum/química , Peptídeos/química , Proinsulina/química , Agregados Proteicos , Proteínas Recombinantes de Fusão/química , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Cromatografia de Afinidade , Corynebacterium glutamicum/genética , Concentração de Íons de Hidrogênio , Peptídeos/genética , Peptídeos/isolamento & purificação , Proinsulina/genética , Proinsulina/isolamento & purificação , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Solubilidade
12.
J Biol Chem ; 293(6): 1908-1909, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29462795

RESUMO

Most newly synthesized proteins destined for the secretory pathway contain a signal peptide (SP) that triggers cotranslational translocation into the endoplasmic reticulum (ER). However, how small polypeptides undergo ER translocation is not fully understood. In this issue of JBC, Guo et al. describe a mechanism for posttranslational translocation of small secretory proteins featuring a positive charge within the SP N-terminal region. Defects in this element disrupt proper secretion and explain the effects of genetic mutations associated with one type of diabetes.


Assuntos
Peptídeos/metabolismo , Proinsulina/química , Sinais Direcionadores de Proteínas , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Peptídeos/química , Peptídeos/genética , Proinsulina/genética , Proinsulina/metabolismo , Biossíntese de Proteínas , Transporte Proteico , Partícula de Reconhecimento de Sinal/genética , Partícula de Reconhecimento de Sinal/metabolismo
13.
Sci Rep ; 6: 22266, 2016 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-26923214

RESUMO

Mycobacterium avium subspecies paratuberculosis (MAP) has been previously associated to T1D as a putative environmental agent triggering or accelerating the disease in Sardinian and Italian populations. Our aim was to investigate the role of MAP in T1D development by evaluating levels of antibodies directed against MAP epitopes and their human homologs corresponding to ZnT8 and proinsulin (PI) in 54 T1D at-risk children from mainland Italy and 42 healthy controls (HCs). A higher prevalence was detected for MAP/ZnT8 pairs (62,96% T1D vs. 7,14% HCs; p < 0.0001) compared to MAP/PI epitopes (22,22% T1D vs. 9,52% HCs) and decreasing trends were observed upon time-point analyses for most peptides. Similarly, classical ZnT8 Abs and GADA decreased in a time-dependent manner, whereas IAA titers increased by 12%. Responses in 0-9 year-old children were stronger than in 10-18 age group (75% vs. 69,1%; p < 0.04). Younger age, female sex and concomitant autoimmune disorders contributed to a stronger seroreactivity suggesting a possible implication of MAP in multiple autoimmune syndrome. Cross-reactivity of the homologous epitopes was reflected by a high correlation coefficient (r(2) > 0.8) and a pairwise overlap of positivity (>83% for MAP/ZnT8).


Assuntos
Proteínas de Transporte de Cátions/imunologia , Diabetes Mellitus Tipo 1/imunologia , Epitopos/imunologia , Mycobacterium avium subsp. paratuberculosis/imunologia , Proinsulina/imunologia , Adolescente , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/química , Antígenos de Bactérias/imunologia , Autoanticorpos/sangue , Autoanticorpos/imunologia , Estudos de Casos e Controles , Proteínas de Transporte de Cátions/química , Criança , Pré-Escolar , Reações Cruzadas/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/microbiologia , Epitopos/química , Feminino , Genótipo , Antígenos HLA/genética , Antígenos HLA/imunologia , Humanos , Lactente , Recém-Nascido , Masculino , Peptídeos/química , Peptídeos/imunologia , Proinsulina/química , Fatores de Risco , Transportador 8 de Zinco
14.
Protein Expr Purif ; 122: 1-7, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26826314

RESUMO

Successfully recovering proinsulin's native conformation from inclusion body is the crucial step to guarantee high efficiency for insulin's manufacture. Here, two by-products of disulfide-linked oligomers and disulfide-isomerized monomers were clearly identified during proinsulin aspart's refolding through multiple analytic methods. Arginine and urea are both used to assist in proinsulin refolding, however the efficacy and possible mechanism was found to be different. The oligomers formed with urea were of larger size than with arginine. With the urea concentrations increasing from 2 M to 4 M, the content of oligomers decreased greatly, but simultaneously the refolding yield at the protein concentration of 0.5 mg/mL decreased from 40% to 30% due to the increase of disulfide-isomerized monomers. In contrast, with arginine concentrations increasing up to 1 M, the refolding yield gradually increased to 50% although the content for oligomers also decreased. Moreover, it was demonstrated that not redox pairs but only oxidant was necessary to facilitate the native disulfide bonds formation for the reduced denatured proinsulin. An oxidative agent of selenocystamine could increase the yield up to 80% in the presence of 0.5 M arginine. Further study demonstrated that refolding with 2 M urea instead of 0.5 M arginine could achieve similar yield as protein concentration is slightly reduced to 0.3 mg/mL. In this case, refolded proinsulin was directly purified through one-step of anionic exchange chromatography, with a recovery of 32% and purity up to 95%. All the results could be easily adopted in insulin's industrial manufacture for improving the production efficiency.


Assuntos
Arginina/química , Cistamina/análogos & derivados , Compostos Organosselênicos/química , Proinsulina/química , Redobramento de Proteína , Ureia/química , Animais , Soluções Tampão , Cistamina/química , Dissulfetos/química , Escherichia coli/química , Escherichia coli/genética , Humanos , Corpos de Inclusão/química , Oxirredução , Proinsulina/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
15.
ACS Chem Biol ; 9(3): 683-91, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24328449

RESUMO

Insulin remains a challenging synthetic target due in large part to its two-chain, disulfide-constrained structure. Biomimetic single chain precursors inspired by proinsulin that utilize short peptides to join the A and B chains can dramatically enhance folding efficiency. Systematic chemical analysis of insulin precursors using an optimized synthetic protocol identified a 49 amino acid peptide named DesDi, which folds with high efficiency by virtue of an optimized structure and could be proteolytically converted to bioactive two-chain insulin. In subsequent applications, we observed that the folding of the DesDi precursor was highly tolerant to amino acid substitution at various insulin residues. The versatility of DesDi as a synthetic insulin precursor was demonstrated through the preparation of several alanine mutants (A10, A16, A18, B12, B15), as well as ValA16, an analog that was unattainable in prior reports. In vitro bioanalysis highlighted the importance of the native, hydrophobic residues at A16 and B15 as part of the core structure of the hormone and revealed the significance of the A18 residue to receptor selectivity. We propose that the DesDi precursor is a versatile synthetic intermediate for the preparation of diverse insulin analogs. It should enable a more comprehensive analysis of function to insulin structure than might not be otherwise possible through conventional approaches.


Assuntos
Insulina/análogos & derivados , Insulina/síntese química , Precursores de Proteínas/química , Alanina/química , Alanina/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Células HEK293 , Humanos , Insulina/química , Insulina/genética , Fator de Crescimento Insulin-Like I/genética , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Proinsulina/química , Dobramento de Proteína , Precursores de Proteínas/genética , Redobramento de Proteína , Transfecção
16.
FEBS Lett ; 587(13): 1942-50, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23669362

RESUMO

Dominant mutations in the human insulin gene can lead to pancreatic ß-cell dysfunction and diabetes mellitus due to toxic folding of a mutant proinsulin. Analogous to a classical mouse model (the Akita mouse), this monogenic syndrome highlights the susceptibility of human ß-cells to endoreticular stress due to protein misfolding and aberrant aggregation. The clinical mutations directly or indirectly perturb native disulfide pairing. Whereas the majority of mutations introduce or remove a cysteine (leading in either case to an unpaired residue), non-cysteine-related mutations identify key determinants of folding efficiency. Studies of such mutations suggest that the evolution of insulin has been constrained not only by its structure and function, but also by the susceptibility of its single-chain precursor to impaired foldability.


Assuntos
Diabetes Mellitus/genética , Proinsulina/genética , Animais , Cistina/química , Cistina/metabolismo , Diabetes Mellitus/metabolismo , Humanos , Modelos Moleculares , Mutação , Proinsulina/química , Proinsulina/metabolismo , Dobramento de Proteína , Estrutura Terciária de Proteína , Deficiências na Proteostase/genética , Deficiências na Proteostase/metabolismo
17.
Biochim Biophys Acta ; 1834(1): 182-90, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22982989

RESUMO

Human M-proinsulin was cleaved by trypsin at the R(31)R(32)-E(33) and K(64)R(65)-G(66) bonds (B/C and C/A junctions), showing the same cleavage specificity as exhibited by prohormone convertases 1 and 2 respectively. Buffalo/bovine M-proinsulin was also cleaved by trypsin at the K(59)R(60)-G(61) bond but at the B/C junction cleavage occurred at the R(31)R(32)-E(33) as well as the R(31)-R(32)E(33) bond. Thus, the human isoform in the native state, with a 31 residue connecting C-peptide, seems to have a unique structure around the B/C and C/A junctions and cleavage at these sites is predominantly governed by the structure of the proinsulin itself. In the case of both the proinsulin species the cleavage at the B/C junction was preferred (65%) over that at the C/A junction (35%) supporting the earlier suggestion of the presence of some form of secondary structure at the C/A junction. Proinsulin and its derivatives, as natural substrates for trypsin, were used and mass spectrometric analysis showed that the k(cat.)/K(m) values for the cleavage were most favourable for the scission of the bonds at the two junctions (1.02±0.08×10(5)s(-1)M(-1)) and the cleavage of the K(29)-T(30) bond of M-insulin-RR (1.3±0.07×10(5)s(-1)M(-1)). However, the K(29)-T(30) bond in M-insulin, insulin as well as M-proinsulin was shielded from attack by trypsin (k(cat.)/K(m) values around 1000s(-1)M(-1)). Hence, as the biosynthetic path follows the sequence; proinsulin→insulin-RR→insulin, the K(29)-T(30) bond becomes shielded, exposed then shielded again respectively.


Assuntos
Peptídeos/química , Proinsulina/química , Tripsina/química , Humanos , Cinética , Espectrometria de Massas , Peptídeos/metabolismo , Proinsulina/metabolismo , Tripsina/metabolismo
18.
J Diabetes Sci Technol ; 6(2): 277-88, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22538136

RESUMO

Insulin is susceptible to thermal fibrillation, a misfolding process that leads to nonnative cross-ß assembly analogous to pathological amyloid deposition. Pharmaceutical formulations are ordinarily protected from such degradation by sequestration of the susceptible monomer within native protein assemblies. With respect to the safety and efficacy of insulin pumps, however, this strategy imposes an intrinsic trade-off between pharmacokinetic goals (rapid absorption and clearance) and the requisite physical properties of a formulation (prolonged shelf life and stability within the reservoir). Available rapid-acting formulations are suboptimal in both respects; susceptibility to fibrillation is exacerbated even as absorption is delayed relative to the ideal specifications of a closed-loop system. To circumvent this molecular trade-off, we exploited structural models of insulin fibrils and amyloidogenic intermediates to define an alternative protective mechanism. Single-chain insulin (SCI) analogs were shown to be refractory to thermal fibrillation with maintenance of biological activity for more than 3 months under conditions that promote the rapid fibrillation and inactivation of insulin. The essential idea exploits an intrinsic incompatibility between SCI topology and the geometry of cross-ß assembly. A peptide tether was thus interposed between the A- and B-chains whose length was (a) sufficiently long to provide the "play" needed for induced fit of the hormone on receptor binding and yet (b) sufficiently short to impose a topological barrier to fibrillation. Our findings suggest that ultrastable monomeric SCI analogs may be formulated without protective self-assembly and so permit simultaneous optimization of pharmacokinetics and reservoir life.


Assuntos
Temperatura Alta , Hipoglicemiantes/química , Sistemas de Infusão de Insulina , Proinsulina/química , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Química Farmacêutica , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/tratamento farmacológico , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/farmacocinética , Masculino , Modelos Moleculares , Proinsulina/administração & dosagem , Proinsulina/farmacocinética , Conformação Proteica , Desnaturação Proteica , Dobramento de Proteína , Estabilidade Proteica , Ratos , Ratos Endogâmicos Lew , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo
19.
J Biol Chem ; 287(1): 43-47, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22105075

RESUMO

For insulin synthesis, the proinsulin precursor is translated at the endoplasmic reticulum (ER), folds to include its three native disulfide bonds, and is exported to secretory granules for processing and secretion. Protein disulfide isomerase (PDI) has long been assumed to assist proinsulin in this process. Herein we have examined the effect of PDI knockdown (PDI-KD) in ß-cells. The data establish that upon PDI-KD, oxidation of proinsulin to form native disulfide bonds is unimpaired and in fact enhanced. This is accompanied by improved proinsulin exit from the ER and increased total insulin secretion, with no evidence of ER stress. We provide evidence for direct physical interaction between PDI and proinsulin in the ER of pancreatic ß-cells, in a manner requiring the catalytic activity of PDI. In ß-cells after PDI-KD, enhanced export is selective for proinsulin over other secretory proteins, but the same effect is observed for recombinant proinsulin trafficking upon PDI-KD in heterologous cells. We hypothesize that PDI exhibits unfoldase activity for proinsulin, increasing retention of proinsulin within the ER of pancreatic ß-cells.


Assuntos
Retículo Endoplasmático/metabolismo , Células Secretoras de Insulina/citologia , Proinsulina/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Sequência de Bases , Dissulfetos/química , Técnicas de Silenciamento de Genes , Células HEK293 , Células Hep G2 , Humanos , Proinsulina/química , Isomerases de Dissulfetos de Proteínas/deficiência , Isomerases de Dissulfetos de Proteínas/genética , Transporte Proteico
20.
J Biol Chem ; 286(51): 43710-43716, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22033917

RESUMO

Mutations in the insulin gene can impair proinsulin folding and cause diabetes mellitus. Although crystal structures of insulin dimers and hexamers are well established, proinsulin is refractory to crystallization. Although an NMR structure of an engineered proinsulin monomer has been reported, structures of the wild-type monomer and hexamer remain undetermined. We have utilized hydroxyl radical footprinting and molecular modeling to characterize these structures. Differences between the footprints of insulin and proinsulin, defining a "shadow" of the connecting (C) domain, were employed to refine the model. Our results demonstrate that in its monomeric form, (i) proinsulin contains a native-like insulin moiety and (ii) the C-domain footprint resides within an adjoining segment (residues B23-B29) that is accessible to modification in insulin but not proinsulin. Corresponding oxidation rates were observed within core insulin moieties of insulin and proinsulin hexamers, suggesting that the proinsulin hexamer retains an A/B structure similar to that of insulin. Further similarities in rates of oxidation between the respective C-domains of proinsulin monomers and hexamers suggest that this loop in each case flexibly projects from an outer surface. Although dimerization or hexamer assembly would not be impaired, an ensemble of predicted C-domain positions would block hexamer-hexamer stacking as visualized in classical crystal lattices. We anticipate that protein footprinting in combination with modeling, as illustrated here, will enable comparative studies of diabetes-associated mutant proinsulins and their aberrant modes of aggregation.


Assuntos
Mutação , Proinsulina/química , Proinsulina/genética , Biologia Computacional/métodos , Cristalografia por Raios X/métodos , Diabetes Mellitus/metabolismo , Dimerização , Radicais Livres , Humanos , Insulina/química , Espectrometria de Massas/métodos , Modelos Moleculares , Peptídeos/química , Conformação Proteica , Estrutura Terciária de Proteína , Software , Solventes/química , Síncrotrons
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA