Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Pathogens ; 9(5)2020 May 14.
Article in English | MEDLINE | ID: mdl-32422907

ABSTRACT

The targeting of immunogens/vaccines to specific immune cells is a promising approach for amplifying immune responses in the absence of exogenous adjuvants. However, the targeting approaches reported thus far require novel, labor-intensive reagents for each vaccine and have primarily been shown as proof-of-concept with isolated proteins and/or inactivated bacteria. We have engineered a plasmid-based, complement receptor-targeting platform that is readily applicable to live forms of multiple gram-negative bacteria, including, but not limited to, Escherichia coli, Klebsiella pneumoniae, and Francisella tularensis. Using F. tularensis as a model, we find that targeted bacteria show increased binding and uptake by macrophages, which coincides with increased p38 and p65 phosphorylation. Mice vaccinated with targeted bacteria produce higher titers of specific antibody that recognizes a greater diversity of bacterial antigens. Following challenge with homologous or heterologous isolates, these mice exhibited less weight loss and/or accelerated weight recovery as compared to counterparts vaccinated with non-targeted immunogens. Collectively, these findings provide proof-of-concept for plasmid-based, complement receptor-targeting of live gram-negative bacteria.

2.
mBio ; 10(4)2019 07 02.
Article in English | MEDLINE | ID: mdl-31266877

ABSTRACT

Fatal outcomes following influenza infection are often associated with secondary bacterial infections. Allergic airway disease (AAD) is known to influence severe complications from respiratory infections, and yet the mechanistic effect of AAD on influenza virus-Streptococcus pneumoniae coinfection has not been investigated previously. We examined the impact of AAD on host susceptibility to viral-bacterial coinfections. We report that AAD improved survival during coinfection when viral-bacterial challenge occurred 1 week after AAD. Counterintuitively, mice with AAD had significantly deceased proinflammatory responses during infection. Specifically, both CD4+ and CD8+ T cell interferon gamma (IFN-γ) responses were suppressed following AAD. Resistance to coinfection was also associated with strong transforming growth factor ß1 (TGF-ß1) expression and increased bacterial clearance. Treatment of AAD mice with IFN-γ or genetic deletion of TGF-ß receptor II expression reversed the protective effects of AAD. Using a novel triple-challenge model system, we show for the first time that AAD can provide protection against influenza virus-S. pneumoniae coinfection through the production of TGF-ß that suppresses the influenza virus-induced IFN-γ response, thereby preserving antibacterial immunity.IMPORTANCE Asthma has become one of the most common chronic diseases and has been identified as a risk factor for developing influenza. However, the impact of asthma on postinfluenza secondary bacterial infection is currently not known. Here, we developed a novel triple-challenge model of allergic airway disease, primary influenza infection, and secondary Streptococcus pneumoniae infection to investigate the impact of asthma on susceptibility to viral-bacterial coinfections. We report for the first time that mice recovering from acute allergic airway disease are highly resistant to influenza-pneumococcal coinfection and that this resistance is due to inhibition of influenza virus-mediated impairment of bacterial clearance. Further characterization of allergic airway disease-associated resistance against postinfluenza secondary bacterial infection may aid in the development of prophylactic and/or therapeutic treatment against coinfection.


Subject(s)
Asthma/complications , Coinfection/immunology , Coinfection/pathology , Disease Susceptibility , Influenza, Human/immunology , Influenza, Human/pathology , Pneumococcal Infections/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Humans , Influenza A virus/growth & development , Interferon-gamma/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Pneumococcal Infections/immunology , Streptococcus pneumoniae/growth & development , Survival Analysis , Transforming Growth Factor beta/metabolism
3.
Infect Immun ; 86(4)2018 04.
Article in English | MEDLINE | ID: mdl-29311236

ABSTRACT

Opsonizing antibody is a critical component of the host protective immune response against many respiratory pathogens. However, the role of antibodies in protection against pulmonary infection with highly virulent Francisella tularensis strain SchuS4 is unclear, and the mechanism that allows F. tularensis to evade antibody-mediated bacterial clearance is not fully understood. We have now found that depletion of alveolar macrophages reveals an otherwise cryptic protective effect of opsonizing antibody. While antibody opsonization alone failed to confer any survival benefit against SchuS4 lung infection, significant protection was observed when mice were depleted of alveolar macrophages prior to infection. Blood immune signature analyses and bacterial burden measurements indicated that the treatment regimen blocked establishment of productive, systemic infection. In addition, protection was found to be dependent upon neutrophils. The results show for the first time a protective effect of opsonizing antibodies against highly virulent F. tularensis SchuS4 pulmonary infection through depletion of alveolar macrophages, the primary bacterial reservoir, and prevention of systemic dissemination. These findings have important implications for the potential use of therapeutic antibodies against intracellular pathogens that may escape clearance by residing within mucosal macrophages.


Subject(s)
Francisella tularensis/immunology , Immunity, Humoral , Macrophages, Alveolar/immunology , Pneumonia/immunology , Pneumonia/microbiology , Tularemia/immunology , Tularemia/microbiology , Animals , Antibodies, Bacterial/immunology , Macrophages, Alveolar/microbiology , Mice , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Respiratory Burst , Sepsis/immunology , Sepsis/microbiology
4.
Infect Immun ; 85(6)2017 06.
Article in English | MEDLINE | ID: mdl-28373354

ABSTRACT

Francisella tularensis causes lethal pneumonia following infection of the lungs by targeting macrophages for intracellular replication; however, macrophages stimulated with interferon gamma (IFN-γ) can resist infection in vitro We therefore hypothesized that the protective effect of IFN-γ against F. tularensisin vivo requires macrophages receptive to stimulation. We found that the lethality of pulmonary F. tularensis LVS infection was exacerbated under conditions of alveolar macrophage depletion and in mice with a macrophage-specific defect in IFN-γ signaling (termed mice with macrophages insensitive to IFN-γ [MIIG mice]). We previously found that treatment with exogenous interleukin 12 (IL-12) protects against F. tularensis infection; this protection was lost in MIIG mice. MIIG mice also exhibited reduced neutrophil recruitment to the lungs following infection. Systemic neutrophil depletion was found to render wild-type mice highly sensitive to respiratory F. tularensis infection, and depletion beginning at 3 days postinfection led to more pronounced sensitivity than depletion beginning prior to infection. Furthermore, IL-12-mediated protection required NADPH oxidase activity. These results indicate that lung macrophages serve a critical protective role in respiratory F. tularensis LVS infection. Macrophages require IFN-γ signaling to mediate protection, which ultimately results in recruitment of neutrophils to further aid in survival from infection.


Subject(s)
Interferon-gamma/immunology , Interleukin-12/pharmacology , Macrophages, Alveolar/immunology , Tularemia/immunology , Animals , Francisella tularensis/pathogenicity , Lung/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Neutrophils/immunology , Pneumonia, Bacterial/immunology
5.
J Clin Endocrinol Metab ; 102(1): 1-5, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27802092

ABSTRACT

Context: The most common genetic cause of permanent neonatal diabetes mellitus is activating mutations in KCNJ11, which can usually be treated using oral sulfonylureas (SUs) instead of insulin injections, although some mutations are SU unresponsive. In this work, we provide a report of the pancreatic islet endocrine cell composition and area in a patient with an SU-unresponsive KCNJ11 mutation (p.G334D), in comparison with age-matched controls. Case Description: Pancreatic autopsy tissue sections from a 2-year-old female child diagnosed with KCNJ11-related diabetes at 4 days of age and 13 age-matched controls were stained with insulin, glucagon, somatostatin, pancreatic polypeptide, and Ki67 antibodies to determine islet endocrine cell composition and area. ß-cell ultrastructure was assessed by electron microscopic (EM) analysis. The patient's pancreas (sampling from head to tail) revealed insulin-positive cells in all regions. The pancreatic ß-cell (insulin) area was significantly reduced compared with controls: 0.50% ± 0.04% versus 1.67% ± 0.20%, respectively (P < 0.00001). There were no significant differences in α-cell (glucagon) or δ-cell (somatostatin) area. EM analysis revealed secretory granules with a dense core typical of mature ß-cells as well as granules with a lighter core characteristic of immature granules. Conclusions: Our results suggest that mechanisms exist that allow preservation of ß-cells in the absence of insulin secretion. It remains to be determined to what extent this reduction in ß-cells may be reversible.


Subject(s)
Diabetes Mellitus/drug therapy , Insulin-Secreting Cells/pathology , Insulin/metabolism , Mutation/genetics , Potassium Channels, Inwardly Rectifying/genetics , Sulfonylurea Compounds/pharmacology , Autopsy , Biomarkers/analysis , Blood Glucose/analysis , Case-Control Studies , Child, Preschool , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Drug Resistance , Female , Humans , Infant , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Prognosis
7.
Proc Natl Acad Sci U S A ; 111(33): E3395-404, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25092300

ABSTRACT

Insulin provides a classical model of a globular protein, yet how the hormone changes conformation to engage its receptor has long been enigmatic. Interest has focused on the C-terminal B-chain segment, critical for protective self-assembly in ß cells and receptor binding at target tissues. Insight may be obtained from truncated "microreceptors" that reconstitute the primary hormone-binding site (α-subunit domains L1 and αCT). We demonstrate that, on microreceptor binding, this segment undergoes concerted hinge-like rotation at its B20-B23 ß-turn, coupling reorientation of Phe(B24) to a 60° rotation of the B25-B28 ß-strand away from the hormone core to lie antiparallel to the receptor's L1-ß2 sheet. Opening of this hinge enables conserved nonpolar side chains (Ile(A2), Val(A3), Val(B12), Phe(B24), and Phe(B25)) to engage the receptor. Restraining the hinge by nonstandard mutagenesis preserves native folding but blocks receptor binding, whereas its engineered opening maintains activity at the price of protein instability and nonnative aggregation. Our findings rationalize properties of clinical mutations in the insulin family and provide a previously unidentified foundation for designing therapeutic analogs. We envisage that a switch between free and receptor-bound conformations of insulin evolved as a solution to conflicting structural determinants of biosynthesis and function.


Subject(s)
Insulin/metabolism , Receptor, Insulin/metabolism , Crystallography, X-Ray , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Binding
8.
Genetics ; 196(2): 539-55, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24281154

ABSTRACT

Drosophila melanogaster has been widely used as a model of human Mendelian disease, but its value in modeling complex disease has received little attention. Fly models of complex disease would enable high-resolution mapping of disease-modifying loci and the identification of novel targets for therapeutic intervention. Here, we describe a fly model of permanent neonatal diabetes mellitus and explore the complexity of this model. The approach involves the transgenic expression of a misfolded mutant of human preproinsulin, hINS(C96Y), which is a cause of permanent neonatal diabetes. When expressed in fly imaginal discs, hINS(C96Y) causes a reduction of adult structures, including the eye, wing, and notum. Eye imaginal discs exhibit defects in both the structure and the arrangement of ommatidia. In the wing, expression of hINS(C96Y) leads to ectopic expression of veins and mechano-sensory organs, indicating disruption of wild-type signaling processes regulating cell fates. These readily measurable "disease" phenotypes are sensitive to temperature, gene dose, and sex. Mutant (but not wild-type) proinsulin expression in the eye imaginal disc induces IRE1-mediated XBP1 alternative splicing, a signal for endoplasmic reticulum stress response activation, and produces global change in gene expression. Mutant hINS transgene tester strains, when crossed to stocks from the Drosophila Genetic Reference Panel, produce F1 adults with a continuous range of disease phenotypes and large broad-sense heritability. Surprisingly, the severity of mutant hINS-induced disease in the eye is not correlated with that in the notum in these crosses, nor with eye reduction phenotypes caused by the expression of two dominant eye mutants acting in two different eye development pathways, Drop (Dr) or Lobe (L), when crossed into the same genetic backgrounds. The tissue specificity of genetic variability for mutant hINS-induced disease has, therefore, its own distinct signature. The genetic dominance of disease-specific phenotypic variability in our model of misfolded human proinsulin makes this approach amenable to genome-wide association study in a simple F1 screen of natural variation.


Subject(s)
Diabetes Mellitus/genetics , Proinsulin/genetics , Animals , Animals, Genetically Modified , Cluster Analysis , Disease Models, Animal , Drosophila melanogaster , Eye/metabolism , Female , Gene Dosage , Gene Expression Profiling , Humans , Male , Mutation , Phenotype , Proinsulin/chemistry , Protein Folding , Quantitative Trait, Heritable , Transcriptome , Transgenes , Wings, Animal/metabolism
9.
J Biol Chem ; 288(40): 29013-23, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23935095

ABSTRACT

Insulin is a major autoantigen in islet autoimmunity and progression to type 1 diabetes. It has been suggested that the insulin B-chain may be critical to insulin autoimmunity in type 1 diabetes. INS-IGF2 consists of the preproinsulin signal peptide, the insulin B-chain, and eight amino acids of the C-peptide in addition to 138 amino acids from the IGF2 gene. We aimed to determine the expression of INS-IGF2 in human pancreatic islets and autoantibodies in newly diagnosed children with type 1 diabetes and controls. INS-IGF2, expressed primarily in beta cells, showed higher levels of expression in islets from normal compared with donors with either type 2 diabetes (p = 0.006) or high HbA1c levels (p < 0.001). INS-IGF2 autoantibody levels were increased in newly diagnosed patients with type 1 diabetes (n = 304) compared with healthy controls (n = 355; p < 0.001). Displacement with cold insulin and INS-IGF2 revealed that more patients than controls had doubly reactive insulin-INS-IGF2 autoantibodies. These data suggest that INS-IGF2, which contains the preproinsulin signal peptide, the B-chain, and eight amino acids of the C-peptide may be an autoantigen in type 1 diabetes. INS-IGF2 and insulin may share autoantibody-binding sites, thus complicating the notion that insulin is the primary autoantigen in type 1 diabetes.


Subject(s)
Autoimmunity/immunology , Insulin/immunology , Islets of Langerhans/immunology , Mutant Chimeric Proteins/immunology , Protein Precursors/immunology , Adolescent , Autoantibodies/blood , Chromosomes, Human, Pair 11/genetics , DNA, Complementary/genetics , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Electrophoresis, Polyacrylamide Gel , Female , Fluorescent Antibody Technique , Gene Expression Regulation , Genome, Human/genetics , Humans , Insulin/blood , Insulin/genetics , Insulin-Like Growth Factor II/genetics , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Middle Aged , Mutant Chimeric Proteins/blood , Oligonucleotide Array Sequence Analysis , Protein Biosynthesis , Protein Precursors/blood , Receptor-Like Protein Tyrosine Phosphatases, Class 8/metabolism , Transcription, Genetic
10.
Nature ; 493(7431): 241-5, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23302862

ABSTRACT

Insulin receptor signalling has a central role in mammalian biology, regulating cellular metabolism, growth, division, differentiation and survival. Insulin resistance contributes to the pathogenesis of type 2 diabetes mellitus and the onset of Alzheimer's disease; aberrant signalling occurs in diverse cancers, exacerbated by cross-talk with the homologous type 1 insulin-like growth factor receptor (IGF1R). Despite more than three decades of investigation, the three-dimensional structure of the insulin-insulin receptor complex has proved elusive, confounded by the complexity of producing the receptor protein. Here we present the first view, to our knowledge, of the interaction of insulin with its primary binding site on the insulin receptor, on the basis of four crystal structures of insulin bound to truncated insulin receptor constructs. The direct interaction of insulin with the first leucine-rich-repeat domain (L1) of insulin receptor is seen to be sparse, the hormone instead engaging the insulin receptor carboxy-terminal α-chain (αCT) segment, which is itself remodelled on the face of L1 upon insulin binding. Contact between insulin and L1 is restricted to insulin B-chain residues. The αCT segment displaces the B-chain C-terminal ß-strand away from the hormone core, revealing the mechanism of a long-proposed conformational switch in insulin upon receptor engagement. This mode of hormone-receptor recognition is novel within the broader family of receptor tyrosine kinases. We support these findings by photo-crosslinking data that place the suggested interactions into the context of the holoreceptor and by isothermal titration calorimetry data that dissect the hormone-insulin receptor interface. Together, our findings provide an explanation for a wealth of biochemical data from the insulin receptor and IGF1R systems relevant to the design of therapeutic insulin analogues.


Subject(s)
Insulin/chemistry , Insulin/metabolism , Receptor, Insulin/chemistry , Receptor, Insulin/metabolism , Animals , Binding Sites , Calorimetry , Cattle , Cell Line , Crystallography, X-Ray , Humans , Leucine/metabolism , Ligands , Models, Molecular , Protein Binding , Protein Structure, Secondary , Reproducibility of Results
11.
Peptides ; 35(1): 42-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22421509

ABSTRACT

Prohormone convertases (PCs) 1 and 2 are the primary endoproteases involved in the post-translational processing of proThyrotropin Releasing Hormone (proTRH) to give rise to TRH and other proposed biologically active non-TRH peptides. Previous evidence suggests that PC1 is responsible for most proTRH cleavage events. Here, we used the PC1 and PC2 knockout (KO) mouse models to examine the effects of PC1 or PC2 loss on proTRH processing. The PC1KO mouse presented a decrease in five proTRH-derived peptides, whereas the PC2KO mouse showed only lesser reduction in three TRH (Gln-His-Pro), TRH-Gly (Gln-His-Pro-Gly), and the short forms preproTRH(178-184) (pFQ(7)) and preproTRH(186-199) (pSE(14)) of pFE(22) (preproTRH(178-199)). Also, PC1KO and not PC2KO showed a decrease in pEH(24) indicating that PC1 is more important in generating this peptide in the mouse, which differs from previous studies using rat proTRH. Furthermore, downstream effects on thyroid hormone levels were evident in PC1KO mice, but not PC2KO mice suggesting that PC1 plays the more critical role in producing bioactive hypophysiotropic TRH. Yet loss of PC1 did not abolish TRH entirely indicating a complementary action for both enzymes in the normal processing of proTRH. We also show that PC2 alone is responsible for catalyzing the conversion of pFE(22) to pFQ(7) and pSE(14), all peptides implicated in regulation of suckling-induced prolactin release. Collectively, results characterize the specific roles of PC1 and PC2 in proTRH processing in vivo.


Subject(s)
Peptide Fragments/biosynthesis , Proprotein Convertase 1/genetics , Proprotein Convertase 2/genetics , Protein Precursors/biosynthesis , Thyrotropin-Releasing Hormone/biosynthesis , Amino Acid Sequence , Animals , Mice , Mice, Knockout , Molecular Sequence Data , Paraventricular Hypothalamic Nucleus/metabolism , Proprotein Convertase 1/deficiency , Proprotein Convertase 2/deficiency , Sequence Homology, Amino Acid , Triiodothyronine/biosynthesis
12.
J Biol Chem ; 287(18): 14703-17, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22396549

ABSTRACT

The proprotein convertase 1/3 is expressed in the regulated secretory pathway of neural and endocrine cells. Its major function is in the post-translational processing and activation of precursor proteins. The PC1/3 knock-out (KO) mouse model has allowed us to elucidate its physiological functions in studies focused primarily on neuroendocrine tissues. However, PC1/3 is also expressed in cells of the immune system, mainly in macrophages. The present study explores the effects of innate immune challenge in the PC1/3 KO mouse. PC1/3 KO mice have an enlarged spleen with marked disorganization of the marginal zone and red pulp. Immunohistochemical studies using various markers demonstrate a depletion of dendritic cells in PC1/3 KO spleens. When challenged with lipopolysaccharide, PC1/3 KO mice are more susceptible to septic shock than wild-type controls or other PC KO mice, such as PC2 and PC7 null mice. Plasma levels of proinflammatory cytokines (IL-6, IL-1ß, and TNF-α) were very significantly elevated in PC1/3 KO mice, consistent with a hypercytokinemia, i.e. indicative of a major systemic uncontrolled inflammatory response or cytokine storm. Peritoneal macrophages isolated from PC1/3 KO mice also demonstrate elevated cytokine secretion when treated with LPS. Electron micrographs show morphological features indicating a prolonged activation of these cells following LPS stimulation. We also present evidence that the proinflammatory T(h)1 pathway is dominant in the PC1/3 KO mouse model. We conclude that aside from its important role in neuroendocrine functions PC1/3 also has an important role in the regulation of the innate immune system, most likely through the regulation of cytokine secretion in macrophages.


Subject(s)
Cytokines/immunology , Gene Expression Regulation, Enzymologic/immunology , Immunity, Innate , Macrophages, Peritoneal/immunology , Proprotein Convertase 1/immunology , Animals , Cytokines/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Immune System Diseases/genetics , Immune System Diseases/immunology , Immune System Diseases/metabolism , Immune System Diseases/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/ultrastructure , Mice , Mice, Knockout , Proprotein Convertase 1/biosynthesis , Proprotein Convertase 1/genetics , Th1 Cells/enzymology , Th1 Cells/metabolism
13.
Methods Mol Biol ; 768: 3-11, 2011.
Article in English | MEDLINE | ID: mdl-21805235

ABSTRACT

Studies of the biosynthesis of insulin in a human insulinoma beginning in 1965 provided the first evidence for a precursor of insulin, the first such prohormone to be identified. Further studies with isolated rat islets then confirmed that the precursor became labeled more rapidly than insulin and later was converted to insulin by a proteolytic processing system located mainly within the secretory granules of the beta cell and was then stored or secreted. The precursor was designated "proinsulin" in 1967 and was isolated and sequenced from beef and pork sources. These structural studies confirmed that the precursor was a single polypeptide chain which began with the B chain of insulin, continued through a connecting segment of 30-35 amino acids and terminated with the A chain. Paired basic residues were identified at the sites of excision of the C-peptide. Human proinsulin and C-peptide were then similarly obtained and sequenced. The human C-peptide assay was developed and provided a useful tool for measuring insulin levels indirectly in diabetics treated with insulin. The discovery of other precursor proteins for a variety of peptide hormones, neuropeptides, or plasma proteins then followed, with all having mainly dibasic cleavage sites for processing. The subsequent discovery of a similar biosynthetic pathway in yeast led to the identification of eukaryotic families of specialized processing subtilisin-like endopeptidases coupled with carboxypeptidase B-like exopeptidases. Most neuroendocrine peptides are processed by two specialized members of this family - PC2 and/or PC1/3 - followed by carboxypeptidase E (CPE). This brief report concentrates mainly on the role of insulin biosynthesis in providing a useful early paradigm of precursor processing in the secretory pathway.


Subject(s)
Insulin/biosynthesis , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational/physiology , Animals , C-Peptide/biosynthesis , Carboxypeptidase H/metabolism , Cattle , History, 20th Century , Humans , Insulin-Secreting Cells/enzymology , Insulinoma/metabolism , Neuropeptides/metabolism , Proinsulin/biosynthesis , Protein Precursors/history , Rats , Saccharomyces cerevisiae , Swine
14.
J Biol Chem ; 286(20): 17399-421, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21454641

ABSTRACT

Insulin is a small but beautifully organized protein with a unique two-chain structure, the first protein to be sequenced. The mechanism of its biosynthesis invited much initial speculation but was finally clarified by the discovery of proinsulin, its single-chain precursor. The rich present-day field of protein precursor processing via post-translational proteolysis within the secretory pathway arose in the early 1970s as an offshoot of studies on insulin biosynthesis, which provided a novel paradigm for the generation of many other small neuroendocrine peptides. Before long, this mechanism was also found to play a role in the production of a much wider spectrum of proteins traversing the secretory pathway (receptors, growth factors, blood-clotting components, and even many viral envelope proteins) occurring in almost all eukaryotic cells. Indeed, yeast provided a key clue in the search for the proprotein convertases, the endoproteases that work along with carboxypeptidases and other modifying enzymes, such as the amidating enzyme complex (PAM), in converting inactive or less active precursor proteins into their fully active peptide products. In this "Reflections" article, I have tried to recount the people and events in my life that led to my involvement first in basic biochemical research and then on to insulin, proinsulin, and many relevant related areas that continue to fascinate and challenge my colleagues and me, as well as many other biomedical scientists today, as diabetes mellitus increasingly threatens human health throughout our contemporary world.


Subject(s)
Biochemistry/history , Insulin/metabolism , Islets of Langerhans/metabolism , Animals , Biochemistry/methods , History, 20th Century , History, 21st Century , Humans , Insulin/history
15.
Islets ; 3(1): 21-34, 2011.
Article in English | MEDLINE | ID: mdl-21278490

ABSTRACT

Diabetes (T1DM and T2DM) is characterized by a deficit in ß-cell mass. A broader understanding of human ß-cell replication mechanism is thus important to increase ß-cell proliferation for future therapeutic interventions. Here, we show that p27 (Kip1), a CDK inhibitor, is expressed abundantly in isolated adult human islets and interacts with various positive cell cycle regulatory proteins including D-type cyclins (D1, D2 and D3) and their kinase partners, CDK4 and CDK6. Also, we see interaction of cyclin E and its kinase partner, CDK2, with p27 suggesting a critical role of p27 as a negative cell cycle regulator in human islets. Our data demonstrate interaction of p27 with GSK-3 in ß-cells and show, employing rodent ß-cells (INS-1), isolated human islets and purified ß-cells derived from human islets, that siRNA-mediated depletion of GSK-3 or p27 or 1-AKP / BIO - mediated GSK-3 inhibition results in increased ß-cell proliferation. We also see reduction of p27 levels following GSK-3 inactivation or depletion. Our data show that serum induction of quiescent INS-1 cells leads to sequential phosphorylation of p27 on its S10 and T187 residues with faster kinetics for S10 corresponding with the decreased levels of p27. Altogether our findings indicate that p27 levels in ß-cells are stabilized by GSK-3 and thus p27 down regulation following GSK-3 depletion / inactivation plays a critical role in promoting ß-cell replication.


Subject(s)
Cell Division , Cyclin-Dependent Kinase Inhibitor p27/genetics , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Insulin-Secreting Cells/metabolism , Adult , Aged , Animals , Cell Division/genetics , Cell Division/physiology , Cell Line , Cyclin-Dependent Kinase Inhibitor p27/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/genetics , Humans , Insulin-Secreting Cells/physiology , Male , Middle Aged , RNA, Small Interfering/pharmacology , Rats
16.
Islets ; 2(5): 308-17, 2010.
Article in English | MEDLINE | ID: mdl-21099329

ABSTRACT

Pancreatic ß-cell response to glucose stimulation is governed by tightly regulated signaling pathways which have not been fully characterized. A screen for novel signaling intermediates identified Pim3 as a glucose-responsive gene in the ß cell, and here, we characterize its role in the regulation of ß-cell function. Pim3 expression in the ß-cell was first observed through microarray analysis on glucose-stimulated murine insulinoma (MIN6) cells where expression was strongly and transiently induced. In the pancreas, Pim3 expression exhibited similar dynamics and was restricted to the ß cell. Perturbation of Pim3 function resulted in enhanced glucose-stimulated insulin secretion, both in MIN6 cells and in isolated islets from Pim3-/- mice, where the augmentation was specifically seen in the second phase of secretion. Consequently, Pim3-/- mice displayed an increased glucose tolerance in vivo. Interestingly, Pim3-/- mice also exhibited increased insulin sensitivity. Glucose stimulation of isolated Pim3-/- islets resulted in increased phosphorylation of ERK1/2, a kinase involved in regulating ß-cell response to glucose. Pim3 was also found to physically interact with SOCS6 and SOCS6 levels were strongly reduced in Pim3-/- islets. Overexpression of SOCS6 inhibited glucose-induced ERK1/2 activation, strongly suggesting that Pim3 regulates ERK1/2 activity through SOCS6. These data reveal that Pim3 is a novel glucose-responsive gene in the ß cell that negatively regulates insulin secretion by inhibiting the activation of ERK1/2, and through its effect on insulin sensitivity, has potentially a more global function in glucose homeostasis.


Subject(s)
Hyperglycemia , Insulin/metabolism , Islets of Langerhans/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Animals , Cell Line , Cell Size , Gene Expression Profiling , Gene Expression Regulation , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Organ Culture Techniques , Organ Specificity , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
17.
Rev Endocr Metab Disord ; 11(3): 205-15, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20938745

ABSTRACT

Over the last decade our insight into the causes of neonatal diabetes has greatly expanded. Neonatal diabetes was once considered a variant of type 1 diabetes that presented early in life. Recent advances in our understanding of this disorder have established that neonatal diabetes is not an autoimmune disease, but rather is a monogenic form of diabetes resulting from mutations in a number of different genes encoding proteins that play a key role in the normal function of the pancreatic beta-cell. Moreover, a correct genetic diagnosis can affect treatment and clinical outcome. This is especially true for patients with mutations in the genes KCNJ11 or ABCC8 that encode the two protein subunits (Kir6.2 and SUR1, respectively) of the ATP-sensitive potassium channel. These patients can be treated with oral sulfonylurea drugs with better glycemic control and quality of life. Recently, mutations in the insulin gene (INS) itself have been identified as another cause of neonatal diabetes. In this article, we review the role of INS mutations in the pathophysiology of neonatal diabetes.


Subject(s)
Diabetes Mellitus, Type 1/congenital , Diabetes Mellitus, Type 1/genetics , Infant, Newborn, Diseases/genetics , Insulin/genetics , Mutation , Amino Acid Sequence , Animals , Congenital Hyperinsulinism/etiology , Congenital Hyperinsulinism/genetics , Humans , Infant, Newborn , Insulin/biosynthesis , Models, Biological , Molecular Biology , Molecular Sequence Data , Mutation/physiology , Proinsulin/genetics
18.
Islets ; 2(3): 135-45, 2010.
Article in English | MEDLINE | ID: mdl-20657742

ABSTRACT

The pancreatic islet displays diverse patterns of endocrine cell arrangement. The prototypic islet, with insulin-secreting beta-cells forming the core surrounded by other endocrine cells in the periphery, is largely based on studies of normal rodent islets. Recent reports on large animals, including humans, show a difference in islet architecture, in which the endocrine cells are randomly distributed throughout the islet. This particular species difference has raised concerns regarding the interpretation of data based on rodent studies to humans. On the other hand, further variations have been reported in marsupials and some nonhuman primates, which possess an inverted ratio of beta-cells to other endocrine cells. This review discusses the striking plasticity of islet architecture and cellular composition among various species including changes in response to metabolic states within a single species. We propose that this plasticity reflects evolutionary acquired adaptation induced by altered physiological conditions, rather than inherent disparities between species.


Subject(s)
Biological Evolution , Islets of Langerhans/anatomy & histology , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Animals , Computer Simulation , Humans , Invertebrates/physiology , Phylogeny , Species Specificity , Vertebrates/physiology
19.
Islets ; 2(3): 149-55, 2010.
Article in English | MEDLINE | ID: mdl-20657753

ABSTRACT

The islet of Langerhans is a highly vascularized micro-organ consisting of not only ß-cells but multiple cell types such as α-, delta-, pancreatic polypeptide- and epsilon-cells that work together to regulate glucose homeostatis. We have recently proposed a new model of the neonatal islet formation in mice by a process of fission following contiguous endocrine cell proliferation in the form of branched cord-like structures in embryos and newborns. There exist large stretches of interconnected islet structures along large blood vessels in the neonatal pancreas, which, upon further development, segregate into smaller fragments (i.e., islets) that eventually become more spherical by internal proliferation as seen in the adult pancreas. α-cells span these elongated islet-like structures in the developing pancreas, which we hypothesize represent sites of fission and facilitate the eventual formation of discrete islets. The α-cells express both prohormone convertase 2 and 1/3 (PC 2 and PC 1/3, respectively), which resulted in the processing of the proglucagon precursor into glucagon-like peptide 1, thereby leading to local production of this important ß-cell growth factor. Furthermore, while α-cells in the adult basically only express PC 2, significant activation of PC 1/3 is also observed in mouse models of insulin resistance such as pregnant, ob/ ob, db/db and prediabetic NOD mice, which may be a common mechanism in proliferating ß-cells. Our study suggests an important role of α-cells for ß-cell proliferation and further for the endocrine cell network within an islet.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Proprotein Convertase 1/metabolism , Regeneration/physiology , Animals , Animals, Newborn , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Embryo, Mammalian , Female , Glucagon-Secreting Cells/physiology , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred NOD , Mice, Obese , Models, Animal , Prediabetic State/metabolism , Prediabetic State/pathology , Pregnancy
20.
J Neurochem ; 114(1): 215-25, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20412386

ABSTRACT

Neuropeptides are produced from larger precursors by limited proteolysis, first by endopeptidases and then by carboxypeptidases. Major endopeptidases required for these cleavages include prohormone convertase (PC) 1/3 and PC2. In this study, quantitative peptidomics analysis was used to characterize the specific role PC1/3 plays in this process. Peptides isolated from hypothalamus, amygdala, and striatum of PC1/3 null mice were compared with those from heterozygous and wild-type mice. Extracts were labeled with stable isotopic tags and fractionated by HPLC, after which relative peptide levels were determined using tandem mass spectrometry. In total, 92 peptides were found, of which 35 were known neuropeptides or related peptides derived from 15 distinct secretory pathway proteins: 7B2, chromogranin A and B, cocaine- and amphetamine-regulated transcript, procholecystokinin, proenkephalin, promelanin concentrating hormone, proneurotensin, propituitary adenylate cyclase-activating peptide, proSAAS, prosomatosatin, provasoactive intestinal peptide, provasopressin, secretogranin III, and VGF. Among the peptides derived from these proteins, approximately 1/3 were decreased in the PC1/3 null mice relative to wild-type mice, approximately 1/3 showed no change, and approximately 1/3 increased in PC1/3 null. Cleavage sites were analyzed in peptides that showed no change or that decreased in PC1/3 mice, and these results were compared with peptides that showed no change or decreased in previous peptidomic studies with PC2 null mice. Analysis of these sites showed that while PC1/3 and PC2 have overlapping substrate preferences, there are particular cleavage site residues that distinguish peptides preferred by each PC.


Subject(s)
Brain/metabolism , Peptides/metabolism , Proprotein Convertase 1/genetics , Amino Acid Sequence , Animals , Mice , Mice, Knockout , Molecular Sequence Data , Proprotein Convertase 2/genetics , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL
...