Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
J Eur Acad Dermatol Venereol ; 36(9): 1456-1466, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35470924

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is a complex disease with variations in severity and healthcare utilization. Examining patient pathways through analyses of longitudinal patient data provides an opportunity to describe real-world clinical patient care and evaluate healthcare access and treatment. OBJECTIVE: To describe longitudinal care pathways including health care management, treatment patterns and disease progression (by proxy measures) in patients with AD. MATERIALS AND METHODS: This was a longitudinal observational study, which used linked data from national and regional healthcare registers in Sweden. Patients with AD were identified through diagnosis in primary or secondary care or by dispensed medications. Descriptive statistics for number of healthcare visits, type of dispensed drug class, rate of - and time to - referral to secondary care and treatment escalation were calculated. RESULTS: A total of 341 866 patients with AD distributed as 197 959 paediatric (age < 12), 36 133 adolescent (age ≥ 12- < 18) and 107 774 adult (age ≥ 18) patients were included in this study. Healthcare visits to primary and secondary care and dispensation of AD-indicated treatments were more common during the year in which managed AD care was initiated. Topical corticosteroids (TCSs) and emollients were the most frequently used treatments across all age cohorts while systemic treatment was uncommon in all age cohorts. Among patients who initiated treatment with TCSs, 18.2% escalated to TCSs with higher potency following the start of managed AD care. CONCLUSIONS: We found that healthcare contacts and use of AD-indicated treatments were concentrated in the year during which managed AD care was initiated and decreased significantly thereafter. Since a significant proportion of patients with AD have flares and persistent AD, our results suggest that patients with AD may be monitored infrequently and are undertreated. There is a need to inform practitioners about adequate treatment options to provide individualized care, in particular for patients with persistent severe AD.


Subject(s)
Dermatitis, Atopic , Dermatologic Agents , Adolescent , Adult , Child , Cohort Studies , Critical Pathways , Dermatitis, Atopic/drug therapy , Dermatologic Agents/therapeutic use , Emollients/therapeutic use , Glucocorticoids/therapeutic use , Humans , Retrospective Studies
2.
Osteoporos Int ; 32(7): 1301-1311, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33411005

ABSTRACT

A novel cost-effectiveness model framework was developed to incorporate the elevated fracture risk associated with a recent fracture and to allow sequential osteoporosis therapies to be evaluated. Treating patients with severe osteoporosis after a recent fracture with a bone-forming agent followed by antiresorptive therapy can be cost-effective compared with antiresorptive therapy alone. Incorporating these novel technical attributes in economic evaluations can support appropriate policy and reimbursement decision-making. PURPOSE: To develop a cost-effectiveness model accommodating increased fracture risk after a recent fracture and treatment sequencing. METHODS: A micro-simulation cost-utility model was developed to accommodate both treatment sequencing and increased risk with recent fracture. The risk of fracture was estimated and simulated using the FRAX® algorithms combined with Swedish registry data on imminent fracture relative risk. In the base-case cost-effectiveness analysis, a sequential treatment starting with a bone-forming agent for 12 months followed by an antiresorptive agent for 48 months initiated immediately after a major osteoporotic fracture (MOF) in a 70-year-old woman with a T-score of 2.5 or less was compared to an antiresorptive treatment alone for 60 months. The model was populated with data relevant for a UK population reflecting a personal social service perspective. RESULTS: The cost per additional quality-adjusted life year (QALY) gained in the base-case setting was estimated at £34,584. Sensitivity analyses revealed the sequential treatment to be cost-saving compared with administering a bone-forming treatment alone. Without simulating an elevated fracture risk immediately after a recent fracture, the cost per QALY changed from £34,584 to £62,184. CONCLUSION: Incorporating imminent fracture risk in economic evaluations has a significant impact on the cost-effectiveness when evaluating fracture prevention treatments in patients with osteoporosis who sustained a recent fracture. Bone-forming treatment followed by antiresorptive therapy can be cost-effective compared to antiresorptive therapy alone depending on treatment acquisition costs.


Subject(s)
Bone Density Conservation Agents , Osteoporosis, Postmenopausal , Osteoporosis , Osteoporotic Fractures , Aged , Bone Density Conservation Agents/therapeutic use , Cost-Benefit Analysis , Female , Humans , Osteoporosis/complications , Osteoporosis/drug therapy , Osteoporotic Fractures/etiology , Osteoporotic Fractures/prevention & control , Quality-Adjusted Life Years , Sweden/epidemiology
3.
Osteoporos Int ; 32(3): 585-594, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33409591

ABSTRACT

Romosozumab is a novel bone-building drug that reduces fracture risk. This health economic analysis indicates that sequential romosozumab-to-alendronate can be a cost-effective treatment option for postmenopausal women with severe osteoporosis at high risk of fracture. PURPOSE: To estimate the cost-effectiveness of sequential treatment with romosozumab followed by alendronate ("romosozumab-to-alendronate") compared with alendronate alone in patients with severe osteoporosis at high risk of fracture in Sweden. METHODS: A microsimulation model with a Markov structure was used to simulate fractures, costs, and quality-adjusted life years (QALYs), for women treated with romosozumab-to-alendronate or alendronate alone. Patients aged 74 years with a recent major osteoporotic fracture (MOF) were followed from the start of treatment until the age of 100 years or death. Treatment with romosozumab for 12 months was followed by alendronate for up to 48 months or alendronate alone with a maximum treatment duration of 60 months. The analysis had a societal perspective. Efficacy of romosozumab and alendronate were derived from phase III randomized controlled trials. Resource use and unit costs were collected from the literature. Cost-effectiveness was estimated using incremental cost-effectiveness ratio (ICER) with QALYs as effectiveness measures. RESULTS: The base case analysis showed that sequential romosozumab-to-alendronate treatment was associated with 0.089 additional QALYs at an additional cost of €3002 compared to alendronate alone, resulting in an ICER of €33,732. At a Swedish reference willingness-to-pay per QALY of €60,000, romosozumab-to-alendronate had a 97.9% probability of being cost-effective against alendronate alone. The results were most sensitive to time horizon, persistence assumptions, patient age, and treatment efficacy. CONCLUSION: The results of this study indicate that sequential romosozumab-to-alendronate can be a cost-effective treatment option for postmenopausal women with severe osteoporosis at high risk of fracture.


Subject(s)
Bone Density Conservation Agents , Osteoporosis, Postmenopausal , Osteoporosis , Aged , Aged, 80 and over , Alendronate/therapeutic use , Antibodies, Monoclonal , Bone Density Conservation Agents/therapeutic use , Cost-Benefit Analysis , Female , Humans , Osteoporosis, Postmenopausal/complications , Osteoporosis, Postmenopausal/drug therapy , Postmenopause , Quality-Adjusted Life Years , Sweden/epidemiology
5.
Prog Mol Biol Transl Sci ; 140: 47-74, 2016.
Article in English | MEDLINE | ID: mdl-27288825

ABSTRACT

PCSK1, encoding prohormone convertase 1/3 (PC1/3), was one of the first genes linked to monogenic early-onset obesity. PC1/3 is a protease involved in the biosynthetic processing of a variety of neuropeptides and prohormones in endocrine tissues. PC1/3 activity is essential for the activating cleavage of many peptide hormone precursors implicated in the regulation of food ingestion, glucose homeostasis, and energy homeostasis, for example, proopiomelanocortin, proinsulin, proglucagon, and proghrelin. A large number of genome-wide association studies in a variety of different populations have now firmly established a link between three PCSK1 polymorphisms frequent in the population and increased risk of obesity. Human subjects with PC1/3 deficiency, a rare autosomal-recessive disorder caused by the presence of loss-of-function mutations in both alleles, are obese and display a complex set of endocrinopathies. Increasing numbers of genetic diagnoses of infants with persistent diarrhea has recently led to the finding of many novel PCSK1 mutations. PCSK1-deficient infants experience severe intestinal malabsorption during the first years of life, requiring controlled nutrition; these children then become hyperphagic, with associated obesity. The biochemical characterization of novel loss-of-function PCSK1 mutations has resulted in the discovery of new pathological mechanisms affecting the cell biology of the endocrine cell beyond simple loss of enzyme activity, for example, dominant-negative effects of certain mutants on wild-type PC1/3 protein, and activation of the cellular unfolded protein response by endoplasmic reticulum-retained mutants. A better understanding of these molecular and cellular pathologies may illuminate possible treatments for the complex endocrinopathy of PCSK1 deficiency, including obesity.


Subject(s)
Obesity/genetics , Obesity/pathology , Polymorphism, Single Nucleotide/genetics , Proprotein Convertase 1/genetics , Humans
6.
Palliat Support Care ; 14(3): 212-24, 2016 06.
Article in English | MEDLINE | ID: mdl-26235481

ABSTRACT

OBJECTIVES: In clinical palliative cancer care, the diversity of patient concerns over time makes information provision a critical issue, the demands of information-seeking patients presenting a challenge to both the communicative and organizational skills of the health provider. This study puts forward a practice model for communication between patients, their family members, and professional health providers during ongoing palliative chemotherapy; a model which supports the providers in enabling person-centered communication. METHOD: A constant comparative analysis adapted to participatory action research was applied. The model was developed step-wise in three interrelated cycles, with results from previous studies from palliative cancer care processed in relation to professional health providers' experience-based clinical knowledge. In doing this, focus group discussions were carried out with providers and patients to develop and revise the model. RESULTS: The Enabling Sense Making model for person-centered communication gave rise to three domains (which are also the major communicative actors in palliative care): the patient, the family, and the provider. These actors were placed in the context of a communicative arena. The three respective domains were built up in different layers discriminating between significant aspects of person-centered communication, from the manifest that is most usually explicated in dialogues, to the latent that tends to be implicitly mediated. SIGNIFICANCE OF RESULTS: The model intends to facilitate timely reorientation of care from curative treatment or rehabilitation to palliation, as well as the introduction of appropriate palliative interventions over time during palliative phases. In this way the model is to be regarded a frame for directing the awareness of the professionals, which focuses on how to communicate and how to consider the patient's way of reasoning. The model could be used as a complement to other strategic initiatives for the advancement of palliative care communication. It needs to be further evaluated in regard to practice evidence.


Subject(s)
Critical Illness/psychology , Palliative Care/psychology , Patient-Centered Care/methods , Professional-Patient Relations , Ambulatory Care/methods , Ambulatory Care/psychology , Communication , Family/psychology , Humans , Neoplasms/complications , Neoplasms/psychology , Outpatients/psychology , Palliative Care/methods , Palliative Care/standards , Qualitative Research
7.
Phytopathology ; 99(4): 344-52, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19271975

ABSTRACT

European mountain ash ringspot-associated virus (EMARAV) was recently characterized from mountain ash (rowan) (Sorbus aucuparia) in Germany. The virus belongs tentatively to family Bunyaviridae but is not closely related to any classified virus. How commonly EMARAV occurs in ringspot disease (EMARSD) affected mountain ash trees was not reported and was investigated here. Virus-specific detection tools such as reverse transcription-polymerase chain reaction and dot blot hybridization using digoxigenin-labeled RNA probes were developed to test 73 mountain ash trees including 16 trees with no virus-like symptoms from 16 districts in Finland and Viipuri, Russia. All trees were infected with EMARAV. Hence, EMARAV is associated with EMARSD and can also cause latent infections in mountain ash. Symptom expression and the variable relative concentrations of viral RNA detected in leaves showed no correlation. Infectious EMARAV was detected also in dormant branches of trees in winter. Subsequently, genetic variability, geographical differentiation, and evolutionary selection pressures were investigated by analyzing RNA3 sequences from 17 isolates. The putative nucleocapsid (NP) gene sequence (944 nucleotides) showed little variability (identities 97 to 99%) and was under strong purifying selection. Amino acid substitutions were detected in two positions at the N terminus and one position at the C terminus of NP in four isolates. The 3' untranslated region (442 nucleotides) was more variable (identities 94 to 99%). Six isolates from a single sampling site exhibited as wide a genetic variability as isolates from sites that were hundreds of kilometers apart and no spatial differentiation of populations of EMARAV was observed.


Subject(s)
Bunyaviridae/isolation & purification , Plant Diseases/virology , RNA, Viral/isolation & purification , Sorbus/virology , Bunyaviridae/genetics , Finland , Genetic Variation , Nucleocapsid Proteins/genetics , Phylogeny , Plant Leaves/virology , Selection, Genetic , Sequence Analysis, RNA
8.
Endocrinology ; 150(3): 1393-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18948394

ABSTRACT

The prohormone convertases (PCs) 1/3 and 2 accomplish the major proteolytic cleavage events in neuroendocrine tissues; each of these convertases has a small associated binding protein that inhibits convertase action in the secretory pathway. The proSAAS protein binds to PC1/3, whereas the 7B2 protein binds to PC2. However, both convertase-binding proteins are more widely expressed than their cognate enzymes, suggesting that they may perform other functions as well. All known mammalian proSAASs are over 85% conserved; thus, identifying functionally important segments has been impossible. Here, we report the first identification of nonmammalian proSAAS molecules, from Xenopus and zebrafish (Danio rerio). Although these two proteins show an overall amino acid sequence identity of only 29 and 30% with mouse proSAAS, two 14-16 residue hydrophobic segments (predicted to form alpha-helices) and two, nine through 11 residue sequences containing basic convertase cleavage sites are highly conserved; therefore, these sequences may be of functional importance. Confidence that these nonmammalian molecules represent authentic proSAAS is supported by the finding that both inhibit mouse PC1/3 with nanomolar inhibition constants; human furin was not inhibited. In vitro, the two proteins were cleaved by PC2 and furin to three or more peptide products. Both zebrafish and Xenopus proSAAS exhibited neural and endocrine distributions, as assessed by in situ and PCR experiments, respectively. In summary, the identification of proSAAS molecules in lower vertebrates provides clues as to functional regions within this widely expressed neuroendocrine protein.


Subject(s)
Neuropeptides/chemistry , Neuropeptides/genetics , Proprotein Convertases/antagonists & inhibitors , Sequence Homology, Amino Acid , Vertebrates/genetics , Amino Acid Sequence , Animals , Conserved Sequence , Embryo, Nonmammalian , Enzyme Inhibitors/metabolism , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data , Neuroendocrine Cells/metabolism , Neurons/metabolism , Neuropeptides/isolation & purification , Neuropeptides/metabolism , Phylogeny , Protein Structure, Secondary/genetics , Xenopus laevis/genetics , Zebrafish/genetics
9.
Oncogene ; 26(38): 5662-8, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-17334394

ABSTRACT

In a genome-wide screen using differential methylation hybridization (DMH), we have identified a CpG island within the 5' region and untranslated first exon of the secretory granule neuroendocrine protein 1 gene (SGNE1/7B2) that showed hypermethylation in medulloblastomas compared to fetal cerebellum. Bisulfite sequencing and combined bisulfite restriction assay were performed to confirm the methylation status of this CpG island in primary medulloblastomas and medulloblastoma cell lines. Hypermethylation was detected in 16/23 (70%) biopsies and 7/8 (87%) medulloblastoma cell lines, but not in non-neoplastic fetal (n=8) cerebellum. Expression of SGNE1 was investigated by semi-quantitative competitive reverse transcription-polymerase chain reaction and found to be significantly downregulated or absent in all, but one primary medulloblastomas and all cell lines compared to fetal cerebellum. After treatment of medulloblastoma cell lines with 5-aza-2'-deoxycytidine, transcription of SGNE1 was restored. No mutation was found in the coding region of SGNE1 by single-strand conformation polymorphism analysis. Reintroduction of SGNE1 into the medulloblastoma cell line D283Med led to a significant growth suppression and reduced colony formation. In summary, we have identified SGNE1 as a novel epigenetically silenced gene in medulloblastomas. Its frequent inactivation, as well as its inhibitory effect on tumor cell proliferation and focus formation strongly argues for a significant role in medulloblastoma development.


Subject(s)
Cerebellar Neoplasms/pathology , DNA Methylation , Medulloblastoma/pathology , Neuroendocrine Secretory Protein 7B2/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/genetics , CpG Islands/genetics , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Down-Regulation/drug effects , Down-Regulation/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medulloblastoma/genetics , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription, Genetic/drug effects
10.
Neuroscience ; 122(2): 437-47, 2003.
Article in English | MEDLINE | ID: mdl-14614908

ABSTRACT

A somatostatin deficit occurs in the cerebral cortex of Alzheimer's disease patients without a major loss in somatostatin-containing neurons. This deficit could be related to a reduction in the rate of proteolytic processing of peptide precursors. Since the two proprotein convertases (PC)1 and PC2 are responsible for the processing of neuropeptide precursors directed to the regulated secretory pathway, we examined whether they are involved first in the proteolytic processing of prosomatostatin in mouse and human brain and secondly in somatostatin defect associated with Alzheimer's disease. By size exclusion chromatography, the cleavage of prosomatostatin to somatostatin-14 is almost totally abolished in the cortex of PC2 null mice, while the proportions of prosomatostatin and somatostatin-28 are increased. By immunohistochemistry, PC1 and PC2 were localized in many neuronal elements in human frontal and temporal cortex. The convertases levels were quantified by Western blot, as well as the protein 7B2 which is required for the production of active PC2. No significant change in PC1 levels was observed in Alzheimer's disease. In contrast, a marked decrease in the ratio of the PC2 precursor to the total enzymatic pool was observed in the frontal cortex of Alzheimer patients. This decrease coincides with an increase in the binding protein 7B2. However, the content and enzymatic activity of the PC2 mature form were similar in Alzheimer patients and controls. Therefore, the cortical somatostatin defect is not due to convertase alteration occuring during Alzheimer's disease. Further studies will be needed to assess the mechanisms involved in somatostatin deficiency in Alzheimer's disease.


Subject(s)
Alzheimer Disease/enzymology , Proprotein Convertase 2/physiology , Protein Precursors/metabolism , Protein Processing, Post-Translational/physiology , Somatostatin/biosynthesis , Somatostatin/deficiency , Somatostatin/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Analysis of Variance , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Female , Humans , Linear Models , Male , Mice , Mice, Knockout , Proprotein Convertase 2/deficiency , Proprotein Convertase 2/genetics , Protein Precursors/genetics , Protein Processing, Post-Translational/genetics , Rats , Rats, Sprague-Dawley , Somatostatin/genetics
11.
Proc Natl Acad Sci U S A ; 99(5): 3087-92, 2002 Mar 05.
Article in English | MEDLINE | ID: mdl-11854475

ABSTRACT

The serine protease prohormone convertase 2 (PC2), principally involved in the processing of polypeptide hormone precursors in neuroendocrine tissues, requires interaction with the neuroendocrine protein 7B2 to generate an enzymatically active form. 7B2 null mice express no PC2 activity and release large quantities of uncleaved ACTH, resulting in a lethal endocrine condition that resembles pituitary Cushing's (Westphal, C. H., Muller, L., Zhou, A., Bonner-Weir, S., Schambelan, M., Steiner, D. F., Lindberg, I. & Leder, P. (1999) Cell 96, 689). Here, we have compared the 7B2 and PC2 null mouse models to determine why the 7B2 null, but not the PC2 null, exhibits a lethal disease state. Both 7B2 and PC2 nulls contained highly elevated pituitary adrenocorticotropic hormone (ACTH); the neurointermediate lobe content of ACTH in 7B2 nulls was 13-fold higher than in WT mice; that of the PC2 null was 65-fold higher. However, circulating ACTH levels were much higher in the 7B2 null than in the PC2 null. Because hypothalamic inhibitory dopaminergic control represents the major influence on intermediate lobe proopiomelanocortin-derived peptide secretion, dopamine levels were measured, and they revealed that 7B2 null pituitaries contained only one-fourth of WT pituitary dopamine. Adrenalectomized 7B2 null animals survived past the usual time of death at 5 weeks; a month after adrenalectomy, they exhibited normal levels of pituitary dopamine, circulating ACTH, and corticosterone. Elevated corticosterone, therefore, seems to play a central role in the lethal phenotype of the 7B2 null, whereas a 7B2-mediated dopaminergic deficiency state may be involved in the actual ACTH hypersecretion phenomenon. Interestingly, adrenalectomized 7B2 nulls also developed unexpectedly severe obesity.


Subject(s)
Adrenal Glands/metabolism , Adrenocorticotropic Hormone/metabolism , Cushing Syndrome/metabolism , Dopamine/metabolism , Nerve Tissue Proteins/physiology , Pituitary Hormones/physiology , Adrenalectomy , Adrenocorticotropic Hormone/genetics , Animals , Cushing Syndrome/etiology , Cushing Syndrome/mortality , Humans , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neuroendocrine Secretory Protein 7B2 , Phenotype , Pituitary Gland/metabolism , Pituitary Gland, Anterior/metabolism , Pituitary Hormones/genetics , Pro-Opiomelanocortin/genetics , Proprotein Convertase 2 , RNA, Messenger , Subtilisins/genetics , Subtilisins/physiology
12.
J Neurochem ; 79(2): 437-44, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11677272

ABSTRACT

The eukaryotic subtilisin prohormone convertase 2 (PC2) is known to require in vivo exposure to the neuroendocrine protein 7B2 in order to produce an enzymatically active species capable of proteolytic action on prohormone substrates. In the present study, we examined the role of the pentabasic site within 27-kDa 7B2 in this process. We prepared two His-tagged recombinant 7B2s by overexpression in bacteria: 7B2-Ser-Ser (SS), with an inactivating mutation in the CT peptide from Lys171-Lys172 (KK) to SS, rendering the CT peptide non-inhibitory; blockade-SS, a double mutant of both the CT peptide as well as of the pentabasic furin cleavage site. These purified proteins were used in a cell-free proPC2 activation assay. Both 7B2-SS as well as blockade-SS were able to facilitate the activation of proPC2 (as judged by efficient production of enzyme activity), suggesting that cleavage at the furin site is not required for 7B2s lacking inhibitory CT peptides. Plasmids encoding proPC2 and various 7B2s were transiently transfected into human embryonic kidney (HEK293) cells and PC2 enzymatic activity and CT forms in each overnight conditioned medium were measured. Cells transfected with proPC2 and wild-type 7B2 secreted CT peptide cleavage products, but cells transfected with proPC2 and the blockade mutant overwhelmingly secreted intact, 27-kDa, blockaded 7B2. Medium obtained from HEK293 cells transfected with proPC2 and either wild-type 7B2, 7B2-SS, or blockade-SS exhibited PC2 activity, but medium from cells expressing the 7B2 blockade mutant did not. We conclude that cleavage at the 7B2 furin consensus site is required to produce PC2 capable of efficient proteolytic inactivation of the CT peptide.


Subject(s)
Nerve Tissue Proteins/physiology , Pituitary Hormones/physiology , Subtilisins/chemistry , Subtilisins/physiology , Animals , Carboxypeptidase H , Carboxypeptidases/pharmacology , Cell Line , Cell Membrane/enzymology , Enzyme Activation/physiology , Furin , Golgi Apparatus/enzymology , Humans , Mutation/physiology , Nerve Tissue Proteins/genetics , Neuroendocrine Secretory Protein 7B2 , Peptide Fragments/physiology , Pituitary Hormones/genetics , Proprotein Convertase 2 , Rats , Subtilisins/metabolism , Time Factors
13.
Biochemistry ; 40(30): 8790-9, 2001 Jul 31.
Article in English | MEDLINE | ID: mdl-11467939

ABSTRACT

The snail Lymnaea stagnalis produces a neuropeptide precursor protein that contains seven Arg-Gly-Asp (RGD) sites. These sites are recognized and cleaved by one or more prohormone convertases in the first processing step to yield mature neuropeptides in the secretory pathway. Conformations of two synthetic RGD-containing peptides derived from the L. stagnalis precursor protein were determined by NMR spectroscopy. The peptides were tested in a platelet aggregation assay for RGD activity and were processed in vitro by PC2 and furin. The native peptide with a proline following the RGD site has minimal structure around the RGD region, does not inhibit platelet aggregation, and is properly processed by the enzymes PC2 and furin. A variant of the native fragment with a serine following the RGD sequence has a significant amount of a reverse turn around the RGD region, is a potent inhibitor of platelet aggregation, and is processed with the same specificity as the native fragment. The large conformational differences between the two peptides provide a molecular mechanism for effects of proline residues following the RGD site and suggest that precursor processing is influenced more by flexibility than by the conformation of the processing site.


Subject(s)
Endopeptidases/metabolism , Neuropeptides/chemistry , Oligopeptides/chemistry , Oligopeptides/metabolism , Protein Precursors/chemistry , Amino Acid Sequence , Animals , Furin , Humans , Hydrolysis , Lymnaea , Molecular Sequence Data , Neuropeptides/metabolism , Nuclear Magnetic Resonance, Biomolecular , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/metabolism , Protein Conformation , Protein Precursors/metabolism , Protein Processing, Post-Translational , Structure-Activity Relationship , Subtilisins/metabolism
14.
J Neurochem ; 76(6): 1833-41, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11259501

ABSTRACT

The conversion of inactive precursor proteins into bioactive neuropeptides and peptide hormones involves regulated secretory proteins such as prohormone convertases PC1 and PC2. The neuroendocrine protein 7B2 represents a specific binding protein for PC2, and the protein proSAAS, which interacts with PC1, exhibits certain structural and functional homologies with 7B2. With the intention of better understanding the physiological role of proSAAS and its derived peptides, we investigated its tissue localization using a new radioimmunoassay (RIA) to a C-terminal proSAAS-derived peptide. Immunoreactivity corresponding to this SAAS-derived peptide is mostly localized to the brain and gut. Analysis of the brain distribution of the proSAAS-derived peptides indicates that the hypothalamus and pituitary are the two richest areas, consistent with the previously described high expression of PC1 in these two areas. In order to investigate the cleavage of proSAAS by prohormone convertases, we incubated recombinant His-tagged proSAAS with recombinant mouse proPC2 or furin, separated the cleavage products using high-pressure gel permeation chromatography and analyzed the products by RIA. Our results indicate that either PC2 or furin can accomplish in vitro rapid removal and efficient internal processing of the C-terminal peptide, exposing the inhibitory hexapeptide to possible further digestion by carboxypeptidases. Finally, we also studied proSAAS processing in the brains of wild-type and PC2 null mice and found that proSAAS is efficiently processed in vivo. Whereas the C-terminal peptide is mostly internally cleaved in wild-type mouse brain, it is not processed as efficiently in the brain of PC2 null mice, suggesting that PC2 is partially responsible for this cleavage in vivo.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Neuropeptides/metabolism , Proprotein Convertase 1 , Protein Precursors/metabolism , Protein Processing, Post-Translational , Subtilisins/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Enzyme Precursors/metabolism , Furin , Mice , Molecular Sequence Data , Neuropeptides/chemistry , Neuropeptides/genetics , Peptide Fragments/chemistry , Proprotein Convertase 2 , Proprotein Convertases , Protein Precursors/chemistry , Protein Precursors/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity , Transfection , Trypsin
15.
J Biol Chem ; 275(50): 39213-22, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-10995742

ABSTRACT

The prohormone convertases (PCs) are synthesized as zymogens whose propeptides contain several multibasic sites. In this study, we investigated the processing of the PC2 propeptide and its function in the regulation of PC2 activity. By using purified pro-PC2 and directed mutagenesis, we found that the propeptide is first cleaved at the multibasic site separating it from the catalytic domain (primary cleavage site); the intact propeptide thus generated is then sequentially processed at two internal sites. Unlike the mechanism described for furin, our mutagenesis studies show that internal cleavage of the propeptide is not required for activation of pro-PC2. In addition, we identified a point mutation in the primary cleavage site that does not prevent the folding nor the processing of the zymogen but nevertheless results in the generation of an inactive PC2 species. These data suggest that the propeptide cleavage site is directly involved in the folding of the catalytic site. By using synthetic peptides, we found that a PC2 propeptide fragment inhibits PC2 activity, and we identified the inhibitory site as the peptide sequence containing basic residues at the extreme carboxyl terminus of the primary cleavage site. Finally, our study supplies information concerning the intracellular fate of a convertase propeptide by providing evidence that the PC2 propeptide is generated and is internally processed within the secretory granules. In agreement with this localization, an internally cleaved propeptide fragment could be released by stimulated secretion.


Subject(s)
Peptides/chemistry , Subtilisins/chemistry , Subtilisins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Brain/metabolism , Catalytic Domain , Cells, Cultured , Chromatography, High Pressure Liquid , Mice , Mice, Knockout , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , Precipitin Tests , Proprotein Convertase 2 , Radioimmunoassay , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Subtilisins/genetics , Time Factors , Transfection , Tumor Cells, Cultured
16.
J Biol Chem ; 275(47): 36741-9, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-10958789

ABSTRACT

The ubiquitous serine endoprotease furin has been implicated in the activation of bacterial toxins and viral glycoproteins as well as in the metastatic progression of certain tumors. Although high molecular mass bioengineered serpin inhibitors have been well characterized, no small nontoxic nanomolar inhibitors have been reported to date. Here we describe the identification of such inhibitors using positional scanning amidated and acetylated synthetic l- and d-hexapeptide combinatorial libraries. The results indicated that l-Arg or l-Lys in all positions generated the most potent inhibitors. However, further investigation revealed that the peptide terminating groups hindered inhibition. Consequently, a series of non-amidated and acetylated polyarginines was synthesized. The most potent inhibitor identified, nona-l-arginine, had a K(i) for furin of 40 nm. The K(i) values for the related convertases PACE4 and prohormone convertase-1 (PC1) were 110 nm and 2.5 microm, respectively. Although nona-l-arginine was cleaved by furin, the major products after a 6-h incubation at 37 degrees C were hexa- and hepta-l-arginines, both of which retained the great majority of their potency and specificity against furin. Hexa-d-arginine was as potent and specific a furin inhibitor as hexa-l-arginine (K(i) values of hexa-d-arginine: 106 nm, 580 nm, and 13.2 microm for furin, PACE4, and PC1, respectively). PC2 was not inhibited by any polyarginine tested; indeed, PC2 showed an increase in activity of up to 140% of the control in the presence of l-polyarginines. Data are also presented that show extended subsite recognition by furin and PC2. Whereas N-terminal acetylation was found to reduce the inhibitory potency of the l-hexapeptide LLRVKR against furin 8-fold, C-terminal amidation reduced the potency < 2-fold. Conversely, N-terminal acetylation increased the potency against PC2 nearly 3-fold, whereas C-terminal amidation of the same peptide increased the potency by a factor of 1.6. Our data indicate that non-acetylated, poly-d-arginine-derived molecules may represent excellent lead compounds for the development of therapeutically useful furin inhibitors.


Subject(s)
Peptides/pharmacology , Subtilisins/antagonists & inhibitors , Animals , Furin , Kinetics , Mice , Peptide Library , Protein Conformation
17.
J Biol Chem ; 275(19): 14667-77, 2000 May 12.
Article in English | MEDLINE | ID: mdl-10799554

ABSTRACT

Prohormone convertases 1 (PC1) and 2 (PC2) are members of a family of subtilisin-like proprotein convertases responsible for proteolytic maturation of a number of different prohormones and proneuropeptides. Although sharing more than 50% homology in their catalytic domains, PC1 and PC2 exhibit differences in substrate specificity and susceptibility to inhibitors. In addition to these differences, PC2, unlike PC1 and other members of the family, specifically binds the neuroendocrine protein 7B2. In order to identify determinants responsible for the specific properties of the PC2 catalytic domain, we compared its primary sequence with that of other PCs. This allowed us to distinguish a PC2-specific sequence at positions 242-248. We constructed two PC2 mutants in which residues 242 and 243 and residues 242-248 were replaced with the corresponding residues of PC1. Studies of in vivo cleavage of proenkephalin, in vivo production of alpha-MSH from proopiomelanocortin, and in vitro cleavage of a PC2-specific artificial substrate by mutant PC2s did not reveal profound alterations. On the other hand, both mutant pro-PC2s exhibited a considerably reduced ability to bind to 21-kDa 7B2. In addition, inhibition of mutant PC2-(242-248) by the potent natural inhibitor 7B2 CT peptide was almost completely abolished. Taken together, our results show that residues 242-248 do not play a significant role in defining the substrate specificity of PC2 but do contribute greatly to binding 7B2 and are critical for inhibition with the 7B2 CT peptide.


Subject(s)
Enzyme Inhibitors/pharmacology , Mutation , Nerve Tissue Proteins/pharmacology , Pituitary Hormones/pharmacology , Subtilisins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Catalytic Domain , Cell Line , Culture Media, Conditioned , DNA Primers , Enkephalins/metabolism , Humans , Hydrolysis , Mice , Molecular Sequence Data , Neuroendocrine Secretory Protein 7B2 , Proprotein Convertase 2 , Protein Binding , Protein Precursors/metabolism , Protein Processing, Post-Translational , Subtilisins/antagonists & inhibitors , Subtilisins/genetics , alpha-MSH/biosynthesis
18.
FEBS Lett ; 473(2): 135-8, 2000 May 12.
Article in English | MEDLINE | ID: mdl-10812060

ABSTRACT

Prohormone convertases (PCs) 1 and 2 are thought to mediate the proteolytic cleavage of many peptide precursors. Endogenous inhibitors of both PC1 and PC2 have now been identified; the 7B2 protein is a nanomolar inhibitor of PC2, while the novel protein proSAAS was recently reported to be a micromolar inhibitor of PC1 [Fricker et al. (2000) J. Neurosci. 20, 639-648]. We here report evidence that 7B2 and proSAAS exhibit several elements of structural and functional homology. Firstly, 26 kDa human, mouse and rat proSAAS, like all vertebrate 7B2s, contain a proline-rich sequence within the first half of the molecule and also contain a C-terminal 40 residue peptide (SAAS CT peptide) separated from the remainder of the protein by a furin consensus sequence. The SAAS CT peptide contains the precise sequence of a hexapeptide previously identified by combinatorial peptide library screening as a potent inhibitor of PC1, and the vast majority of the inhibitory potency of proSAAS can be attributed to this hexapeptide. Further, like the 7B2 CT peptide, SAAS CT-derived peptides represent tight-binding competitive convertase inhibitors with nanomolar potencies. Lastly, recombinant PC1 is able to cleave the proSAAS CT peptide to a product with a mass consistent with cleavage following the inhibitory hexapeptide. Taken together, our results indicate that proSAAS and 7B2 may comprise two members of a functionally homologous family of convertase inhibitor proteins.


Subject(s)
Aspartic Acid Endopeptidases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Nerve Tissue Proteins/metabolism , Neuropeptides/pharmacology , Pituitary Hormones/metabolism , Proprotein Convertase 1 , Protein Precursors/pharmacology , Amino Acid Sequence , Animals , Aspartic Acid Endopeptidases/metabolism , Binding, Competitive , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Furin , Kinetics , Mice , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Neuroendocrine Secretory Protein 7B2 , Neuropeptides/chemistry , Neuropeptides/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Pituitary Hormones/chemistry , Proprotein Convertase 2 , Proprotein Convertases , Protein Binding , Protein Precursors/chemistry , Protein Precursors/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Subtilisins/antagonists & inhibitors , Subtilisins/metabolism
19.
J Biol Chem ; 275(23): 17886-93, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10749852

ABSTRACT

The prohormone convertases (PCs) are an evolutionarily ancient group of proteases required for the maturation of neuropeptide and peptide hormone precursors. In Drosophila melanogaster, the homolog of prohormone convertase 2, dPC2 (amontillado), is required for normal hatching behavior, and immunoblotting data indicate that flies express 80- and 75-kDa forms of this protein. Because mouse PC2 (mPC2) requires 7B2, a helper protein for productive maturation, we searched the fly data base for the 7B2 signature motif PPNPCP and identified an expressed sequence tag clone encoding the entire open reading frame for this protein. dPC2 and d7B2 cDNAs were subcloned into expression vectors for transfection into HEK-293 cells; mPC2 and rat 7B2 were used as controls. Although active mPC2 was detected in medium in the presence of either d7B2 or r7B2, dPC2 showed no proteolytic activity upon coexpression of either d7B2 or r7B2. Labeling experiments showed that dPC2 was synthesized but not secreted from HEK-293 cells. However, when dPC2 and either d7B2 or r7B2 were coexpressed in Drosophila S2 cells, abundant immunoreactive dPC2 was secreted into the medium, coincident with the appearance of PC2 activity. Expression and secretion of dPC2 enzyme activity thus appears to require insect cell-specific posttranslational processing events. The significant differences in the cell biology of the insect and mammalian enzymes, with 7B2 absolutely required for secretion of dPC2 and zymogen conversion occurring intracellularly in the case of dPC2 but not mPC2, support the idea that the Drosophila enzyme has specific requirements for maturation and secretion that can be met only in insect cells.


Subject(s)
Drosophila melanogaster/enzymology , Nerve Tissue Proteins/metabolism , Pituitary Hormones/metabolism , Subtilisins/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cell Line , Cricetinae , Drosophila melanogaster/genetics , Golgi Apparatus/enzymology , Humans , Kinetics , Mice , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neuroendocrine Secretory Protein 7B2 , Pituitary Hormones/chemistry , Pituitary Hormones/genetics , Polymerase Chain Reaction , Proprotein Convertase 2 , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Subtilisins/chemistry , Subtilisins/genetics , Transfection
20.
Biochem Biophys Res Commun ; 267(3): 940-2, 2000 Jan 27.
Article in English | MEDLINE | ID: mdl-10673395

ABSTRACT

Prohormone convertases play important roles in the proteolytic conversion of many protein precursors. The neuroendocrine protein 7B2 and its 31-residue carboxyl-terminal (CT) peptide potently and specifically inhibit prohormone convertase 2 (PC2). We have analyzed the residues contributing to inhibition using N-terminal truncation and alanine scanning. Removal of more than 3 residues from the amino-terminal end of CT1-18 resulted in a more than 190-fold drop in inhibitory activity, showing that most of the residues between 3 and 18 are required for inhibition. In agreement, an Ala scan indicated that only 4 residues could be replaced with Ala without losing mid-nanomolar inhibitory potency; in particular, Gln7, Gln9, and Asp12 could be Ala-substituted to yield peptides with a similar inhibitory potency to the starting peptide. The all-d-retro-inverso, all-l-inverso, and all-d analogues of CT peptide were completely inactive, indicating that amino acid side chains and the CT peptide main chain interact with PC2. CT peptide inhibition could not be competitively blocked by preincubation with truncated CT peptide forms, supporting an absolute requirement for the Lys-Lys pair in initial binding of the CT peptide to the active site.


Subject(s)
Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Peptide Fragments/pharmacology , Pituitary Hormones/chemistry , Pituitary Hormones/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Mice , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Neuroendocrine Secretory Protein 7B2 , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Proprotein Convertase 2 , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sequence Deletion , Subtilisins/antagonists & inhibitors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...