Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 951
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Biochem ; 83: 249-73, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24905783

RESUMO

Proteolytic enzymes are key signaling molecules in both normal physiological processes and various diseases. After synthesis, protease activity is tightly controlled. Consequently, levels of protease messenger RNA and protein often are not good indicators of total protease activity. To more accurately assign function to new proteases, investigators require methods that can be used to detect and quantify proteolysis. In this review, we describe basic principles, recent advances, and applications of biochemical methods to track protease activity, with an emphasis on the use of activity-based probes (ABPs) to detect protease activity. We describe ABP design principles and use case studies to illustrate the application of ABPs to protease enzymology, discovery and development of protease-targeted drugs, and detection and validation of proteases as biomarkers.


Assuntos
Peptídeo Hidrolases/química , Peptídeo Hidrolases/fisiologia , Animais , Bioquímica/métodos , Biomarcadores/química , Caspases/química , Química Farmacêutica/métodos , Desenho de Fármacos , Escherichia coli/enzimologia , Humanos , Peptídeos/química , Proteômica/métodos , Especificidade por Substrato
2.
Nat Rev Mol Cell Biol ; 16(8): 455-60, 2015 08.
Artigo em Inglês | MEDLINE | ID: mdl-26130008

RESUMO

DNA-protein crosslinks (DPCs) are highly toxic DNA adducts, but whether dedicated DPC-repair mechanisms exist was until recently unknown. This has changed with discoveries made in yeast and Xenopus laevis that revealed a protease-based DNA-repair pathway specific for DPCs. Importantly, mutations in the gene encoding the putative human homologue of a yeast DPC protease cause a human premature ageing and cancer predisposition syndrome. Thus, DPC repair is a previously overlooked genome-maintenance mechanism that may be essential for tumour suppression.


Assuntos
Adutos de DNA/genética , Reparo do DNA , Animais , Adutos de DNA/metabolismo , Instabilidade Genômica , Humanos , Peptídeo Hidrolases/fisiologia
3.
Proc Natl Acad Sci U S A ; 119(33): e2202661119, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35939668

RESUMO

In Staphylococcus aureus, virulence is under the control of a quorum sensing (QS) circuit encoded in the accessory gene regulator (agr) genomic locus. Key to this pathogenic behavior is the production and signaling activity of a secreted pheromone, the autoinducing peptide (AIP), generated following the ribosomal synthesis and posttranslational modification of a precursor polypeptide, AgrD, through two discrete cleavage steps. The integral membrane protease AgrB is known to catalyze the first processing event, generating the AIP biosynthetic intermediate, AgrD (1-32) thiolactone. However, the identity of the second protease in this biosynthetic pathway, which removes an N-terminal leader sequence, has remained ambiguous. Here, we show that membrane protease regulator of agr QS (MroQ), an integral membrane protease recently implicated in the agr response, is directly involved in AIP production. Genetic complementation and biochemical experiments reveal that MroQ proteolytic activity is required for AIP biosynthesis in agr specificity group I and group II, but not group III. Notably, as part of this effort, the biosynthesis and AIP-sensing arms of the QS circuit were reconstituted together in vitro. Our experiments also reveal the molecular features guiding MroQ cleavage activity, a critical factor in defining agr specificity group identity. Collectively, our study adds to the molecular understanding of the agr response and Staphylococcus aureus virulence.


Assuntos
Proteínas de Bactérias , Proteínas de Membrana , Peptídeo Hidrolases , Feromônios , Percepção de Quorum , Staphylococcus aureus , Transativadores , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/fisiologia , Proteínas de Membrana/fisiologia , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/fisiologia , Feromônios/biossíntese , Percepção de Quorum/genética , Staphylococcus aureus/patogenicidade , Transativadores/genética , Transativadores/metabolismo , Virulência
4.
Mol Microbiol ; 115(2): 290-304, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32996200

RESUMO

Bacteroides fragilis toxin (BFT) is a protein secreted by enterotoxigenic (ETBF) strains of B. fragilis. BFT is synthesized as a proprotein (proBFT) that is predicted to be a lipoprotein and that is cleaved into two discrete fragments by a clostripain-like protease called fragipain (Fpn). In this study, we obtained evidence that Fpn cleaves proBFT following its transport across the outer membrane. Remarkably, we also found that the disruption of the fpn gene led to a strong reduction in the level of >100 other proteins, many of which are predicted to be lipoproteins, in the culture medium of an ETBF strain. Experiments performed with purified Fpn provided direct evidence that the protease releases at least some of these proteins from the cell surface. The observation that wild-type cells outcompeted an fpn- strain in co-cultivation assays also supported the notion that Fpn plays an important role in cell physiology and is not simply dedicated to toxin biogenesis. Finally, we found that purified Fpn altered the adhesive properties of HT29 intestinal epithelial cells. Our results suggest that Fpn is a broad-spectrum protease that not only catalyzes the protein secretion on a wide scale but that also potentially cleaves host cell proteins during colonization.


Assuntos
Toxinas Bacterianas/metabolismo , Bacteroides fragilis/metabolismo , Metaloendopeptidases/metabolismo , Peptídeo Hidrolases/metabolismo , Bacteroides fragilis/genética , Cisteína Endopeptidases/metabolismo , Lipoproteínas/metabolismo , Peptídeo Hidrolases/fisiologia
5.
PLoS Pathog ; 15(9): e1008029, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31545853

RESUMO

Although Escherichia coli Nissle 1917 (EcN) has been used therapeutically for over a century, the determinants of its probiotic properties remain elusive. EcN produces two siderophore-microcins (Mcc) responsible for an antagonistic activity against other Enterobacteriaceae. EcN also synthesizes the genotoxin colibactin encoded by the pks island. Colibactin is a virulence factor and a putative pro-carcinogenic compound. Therefore, we aimed to decouple the antagonistic activity of EcN from its genotoxic activity. We demonstrated that the pks-encoded ClbP, the peptidase that activates colibactin, is required for the antagonistic activity of EcN. The analysis of a series of ClbP mutants revealed that this activity is linked to the transmembrane helices of ClbP and not the periplasmic peptidase domain, indicating the transmembrane domain is involved in some aspect of Mcc biosynthesis or secretion. A single amino acid substitution in ClbP inactivates the genotoxic activity but maintains the antagonistic activity. In an in vivo salmonellosis model, this point mutant reduced the clinical signs and the fecal shedding of Salmonella similarly to the wild type strain, whereas the clbP deletion mutant could neither protect nor outcompete the pathogen. The ClbP-dependent antibacterial effect was also observed in vitro with other E. coli strains that carry both a truncated form of the Mcc gene cluster and the pks island. In such strains, siderophore-Mcc synthesis also required the glucosyltransferase IroB involved in salmochelin production. This interplay between colibactin, salmochelin, and siderophore-Mcc biosynthetic pathways suggests that these genomic islands were co-selected and played a role in the evolution of E. coli from phylogroup B2. This co-evolution observed in EcN illustrates the fine margin between pathogenicity and probiotic activity, and the need to address both the effectiveness and safety of probiotics. Decoupling the antagonistic from the genotoxic activity by specifically inactivating ClbP peptidase domain opens the way to the safe use of EcN.


Assuntos
Escherichia coli/fisiologia , Mutagênicos/toxicidade , Probióticos/uso terapêutico , Animais , Antibiose/genética , Antibiose/fisiologia , Bacteriocinas/genética , Bacteriocinas/metabolismo , Bacteriocinas/toxicidade , Vias Biossintéticas/genética , Enterobactina/análogos & derivados , Enterobactina/genética , Enterobactina/fisiologia , Enterobactina/toxicidade , Escherichia coli/genética , Escherichia coli/patogenicidade , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Feminino , Genes Bacterianos , Ilhas Genômicas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Família Multigênica , Mutação , Peptídeo Hidrolases/química , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/fisiologia , Peptídeos/genética , Peptídeos/fisiologia , Peptídeos/toxicidade , Policetídeos/toxicidade , Probióticos/toxicidade , Domínios Proteicos , Salmonelose Animal/microbiologia , Salmonelose Animal/terapia , Salmonella typhimurium , Sideróforos/genética , Sideróforos/fisiologia , Sideróforos/toxicidade , Fatores de Virulência/genética , Fatores de Virulência/fisiologia , Fatores de Virulência/toxicidade
6.
J Exp Bot ; 72(9): 3352-3366, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33587751

RESUMO

Plants are a primary food source and can form the basis for renewable energy resources. The final size of their organs is by far the most important trait to consider when seeking increased plant productivity. Being multicellular organisms, plant organ size is mainly determined by the coordination between cell proliferation and cell expansion. The protease DA1 limits the duration of cell proliferation and thereby restricts final organ size. Since its initial identification as a negative regulator of organ growth, various transcriptional regulators of DA1, but also interacting proteins, have been identified. These interactors include cleavage substrates of DA1, and also proteins that modulate the activity of DA1 through post-translational modifications, such as ubiquitination, deubiquitination, and phosphorylation. In addition, many players in the DA1 pathway display conserved phenotypes in other dicot and even monocot species. In this review, we provide a timely overview of the complex, but intriguing, molecular mechanisms that fine-tune the activity of DA1 and therefore final organ size. Moreover, we lay out a roadmap to identify and characterize substrates of proteases and frame the substrate cleavage events in their biological context.


Assuntos
Peptídeo Hidrolases/fisiologia , Proteínas de Plantas/fisiologia , Plantas/enzimologia , Processamento de Proteína Pós-Traducional , Regulação da Expressão Gênica de Plantas
7.
Br J Cancer ; 123(7): 1164-1177, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32636467

RESUMO

BACKGROUND: Deubiquitinating enzymes (DUBs) are linked to cancer progression and dissemination, yet less is known about their regulation and impact on epithelial-mesenchymal transition (EMT). METHODS: An integrative translational approach combining systematic computational analyses of The Cancer Genome Atlas cancer cohorts with CRISPR genetics, biochemistry and immunohistochemistry methodologies to identify and assess the role of human DUBs in EMT. RESULTS: We identify a previously undiscovered biological function of STAM-binding protein like 1 (STAMBPL1) deubiquitinase in the EMT process in lung and breast carcinomas. We show that STAMBPL1 expression can be regulated by mutant p53 and that its catalytic activity is required to affect the transcription factor SNAI1. Accordingly, genetic depletion and CRISPR-mediated gene knockout of STAMBPL1 leads to marked recovery of epithelial markers, SNAI1 destabilisation and impaired migratory capacity of cancer cells. Reversely, STAMBPL1 expression reprogrammes cells towards a mesenchymal phenotype. A significant STAMBPL1-SNAI1 co-signature was observed across multiple tumour types. Importantly, STAMBPL1 is highly expressed in metastatic tissues compared to matched primary tumour of the same lung cancer patient and its expression predicts poor prognosis. CONCLUSIONS: Our study provides a novel concept of oncogenic regulation of a DUB and presents a new role and predictive value of STAMBPL1 in the EMT process across multiple carcinomas.


Assuntos
Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal , Neoplasias Pulmonares/patologia , Peptídeo Hidrolases/fisiologia , Linhagem Celular Tumoral , Enzimas Desubiquitinantes/fisiologia , Feminino , Humanos , Peptídeo Hidrolases/análise , Fatores de Transcrição da Família Snail/análise , Fatores de Transcrição da Família Snail/fisiologia , Proteína Supressora de Tumor p53/genética
8.
Biochem Biophys Res Commun ; 533(3): 548-552, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-32977947

RESUMO

Wnt/ß-catenin signaling is important for development and progression of colorectal cancer (CRC). The degradation complex for ß-catenin is functionally impaired in CRC cells, thereby resulting in the accumulation of ß-catenin and its translocation into the nucleus. Nuclear ß-catenin interacts with and co-activates T cell factor4 (TCF4), resulting in ß-catenin/TCF4-dependent transcription. Therefore, nuclear ß-catenin has been categorized as the main driving force in the tumorigenesis of CRC. Recent studies reveal that Jun activation domain-binding protein 1 (JAB1) enhances the degradation of seven in absentia homolog-1 (SIAH-1), a putative E3 ubiquitin ligase of ß-catenin, and positively regulates the expression of total ß-catenin in human CRC cells. An another recent study also shows that nuclear ß-catenin is ubiquitinated and degraded by an E3 ubiquitin ligase, tripartite motif-containing protein 33 (TRIM33). However, the regulatory mechanism for the expression of nuclear ß-catenin remains to be fully understood. In this study, we have demonstrated that JAB1 positively regulates the expression of nuclear ß-catenin, c-MYC as a ß-catenin/TCF4 target, and cell cycle regulators, such as Ki-67 and topoisomerase IIα, in human CRC cells. Taken together, these results suggest that JAB1 is considered as a promising target for novel CRC therapy.


Assuntos
Complexo do Signalossomo COP9/fisiologia , Neoplasias Colorretais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Peptídeo Hidrolases/fisiologia , beta Catenina/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo
9.
Nat Immunol ; 9(3): 263-71, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18223652

RESUMO

The paracaspase MALT1 mediates T cell antigen receptor-induced signaling to the transcription factor NF-kappaB and is indispensable for T cell activation and proliferation. Enhanced expression of MALT1 or aberrant expression of a fusion protein of the apoptosis inhibitor API2 and MALT1 has been linked to mucosa-associated lymphoid tissue lymphoma. Despite the presence of a caspase-like domain, MALT1 proteolytic activity has not yet been demonstrated. Here we show that T cell antigen receptor stimulation induced recruitment of the NF-kappaB inhibitor A20 into a complex of MALT1 and the adaptor protein Bcl-10, leading to MALT1-mediated processing of A20. API2-MALT1 expression likewise resulted in cleavage of A20. MALT1 cleaved human A20 after arginine 439 and impaired its NF-kappaB-inhibitory function. Our studies identify A20 as a substrate of MALT1 and emphasize the importance of MALT1 proteolytic activity in the 'fine tuning' of T cell antigen receptor signaling.


Assuntos
Caspases/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , NF-kappa B/antagonistas & inibidores , Proteínas de Neoplasias/fisiologia , Proteínas Nucleares/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Caspases/genética , Linhagem Celular , Proteínas de Ligação a DNA , Humanos , Immunoblotting , Células Jurkat , Ativação Linfocitária/imunologia , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa , Proteínas de Neoplasias/genética , Peptídeo Hidrolases/fisiologia , Transdução de Sinais/imunologia , Transfecção , Proteína 3 Induzida por Fator de Necrose Tumoral alfa
10.
Int J Mol Sci ; 21(18)2020 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-32948029

RESUMO

The crucial role of extracellular proteases in cancer progression is well-known, especially in relation to the promotion of cell invasion through extracellular matrix remodeling. This also occurs by the ability of extracellular proteases to induce the shedding of transmembrane proteins at the plasma membrane surface or within extracellular vesicles. This process results in the regulation of key signaling pathways by the modulation of kinases, e.g., the epidermal growth factor receptor (EGFR). Considering their regulatory roles in cancer, therapeutics targeting various extracellular proteases have been discovered. These include the metal-binding agents di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), which increase c-MET degradation by multiple mechanisms. Both the direct and indirect inhibition of protease expression and activity can be achieved through metal ion depletion. Considering direct mechanisms, chelators can bind zinc(II) that plays a catalytic role in enzyme activity. In terms of indirect mechanisms, Dp44mT and DpC potently suppress the expression of the kallikrein-related peptidase-a prostate-specific antigen-in prostate cancer cells. The mechanism of this activity involves promotion of the degradation of the androgen receptor. Additional suppressive mechanisms of Dp44mT and DpC on matrix metalloproteases (MMPs) relate to their ability to up-regulate the metastasis suppressors N-myc downstream regulated gene-1 (NDRG1) and NDRG2, which down-regulate MMPs that are crucial for cancer cell invasion.


Assuntos
Antineoplásicos/uso terapêutico , Quelantes/uso terapêutico , Ferro , Proteínas de Neoplasias/fisiologia , Peptídeo Hidrolases/fisiologia , Inibidores de Proteases/uso terapêutico , Zinco , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Quelantes/farmacologia , Progressão da Doença , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Líquido Extracelular/enzimologia , Vesículas Extracelulares/enzimologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Ácidos Hidroxâmicos/uso terapêutico , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Calicreínas/antagonistas & inibidores , Calicreínas/fisiologia , Metaloproteinases da Matriz/fisiologia , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Oxaprozina/farmacologia , Oxaprozina/uso terapêutico , Fenilalanina/análogos & derivados , Fenilalanina/farmacologia , Fenilalanina/uso terapêutico , Inibidores de Proteases/farmacologia , Proteínas Quinases/fisiologia , Piridinas/farmacologia , Piridinas/uso terapêutico , Tiofenos/farmacologia , Tiofenos/uso terapêutico , Tiossemicarbazonas/farmacologia , Tiossemicarbazonas/uso terapêutico
11.
J Biol Chem ; 293(11): 3965-3980, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29352108

RESUMO

Protein SUMOylation has been reported to play a role in innate immune response, but the enzymes, substrates, and consequences of the specific inflammatory signaling events are largely unknown. Reactive oxygen species (ROS) are abundantly produced during macrophage activation and required for Toll-like receptor 4 (TLR4)-mediated inflammatory signaling. Previously, we demonstrated that SENP3 is a redox-sensitive SUMO2/3 protease. To explore any links between reversible SUMOylation and ROS-related inflammatory signaling in macrophage activation, we generated mice with Senp3 conditional knock-out in myeloid cells. In bacterial lipopolysaccharide (LPS)-induced in vitro and in vivo inflammation models, we found that SENP3 deficiency markedly compromises the activation of TLR4 inflammatory signaling and the production of proinflammatory cytokines in macrophages exposed to LPS. Moreover, Senp3 conditional knock-out mice were significantly less susceptible to septic shock. Of note, SENP3 deficiency was associated with impairment in JNK phosphorylation. We found that MKK7, which selectively phosphorylates JNK, is a SENP3 substrate and that SENP3-mediated deSUMOylation of MKK7 may favor its binding to JNK. Importantly, ROS-dependent SENP3 accumulation and MKK7 deSUMOylation rapidly occurred after LPS stimulation. In conclusion, our findings indicate that SENP3 potentiates LPS-induced TLR4 signaling via deSUMOylation of MKK7 leading to enhancement in JNK phosphorylation and the downstream events. Therefore this work provides novel mechanistic insights into redox regulation of innate immune responses.


Assuntos
Imunidade Inata/imunologia , Inflamação/patologia , Lipopolissacarídeos/toxicidade , MAP Quinase Quinase 7/metabolismo , Macrófagos/patologia , Peptídeo Hidrolases/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Animais , Células Cultivadas , Cisteína Endopeptidases , Citocinas/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , MAP Quinase Quinase 7/genética , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Sumoilação
12.
J Cell Sci ; 130(22): 3801-3808, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28972134

RESUMO

Cell motility is required for diverse processes during immunity and inflammation. Classically, leukocyte motility is defined as an amoeboid type of migration, however some leukocytes, like macrophages, also employ a more mesenchymal mode of migration. Here, we sought to characterize the mechanisms that regulate neutrophil and macrophage migration in vivo by using real-time imaging of leukocyte motility within interstitial tissues in zebrafish larvae. Neutrophils displayed a rounded morphology and rapid protease-independent motility, lacked defined paxillin puncta, and had persistent rearward polarization of stable F-actin and the microtubule network. By contrast, macrophages displayed an elongated morphology with reduced speed and increased directional persistence and formed paxillin-containing puncta but had a less-defined polarization of the microtubule and actin networks. We also observed differential effects of protease inhibition, microtubule disruption and ROCK inhibition on the efficiency of neutrophil and macrophage motility. Taken together, our findings suggest that larval zebrafish neutrophils and macrophage display distinct modes of migration within interstitial tissues in vivo.


Assuntos
Macrófagos/fisiologia , Neutrófilos/fisiologia , Animais , Movimento Celular , Polaridade Celular , Forma Celular , Rastreamento de Células , Larva/citologia , Microscopia de Fluorescência , Microscopia de Vídeo , Microtúbulos/metabolismo , Paxilina/fisiologia , Peptídeo Hidrolases/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/fisiologia
13.
Med Res Rev ; 38(4): 1295-1331, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29149530

RESUMO

There are numerous proteases of pathogenic organisms that are currently targeted for therapeutic intervention along with many that are seen as potential drug targets. This review discusses the chemical and biological makeup of some key druggable proteases expressed by the five major classes of disease causing agents, namely bacteria, viruses, fungi, eukaryotes, and prions. While a few of these enzymes including HIV protease and HCV NS3-4A protease have been targeted to a clinically useful level, a number are yet to yield any clinical outcomes in terms of antimicrobial therapy. A significant aspect of this review discusses the chemical and pharmacological characteristics of inhibitors of the various proteases discussed. A total of 25 inhibitors have been considered potent and safe enough to be trialed in humans and are at different levels of clinical application. We assess the mechanism of action and clinical performance of the protease inhibitors against infectious agents with their developmental strategies and look to the next frontiers in the use of protease inhibitors as anti-infective agents.


Assuntos
Doenças Transmissíveis/tratamento farmacológico , Peptídeo Hidrolases/fisiologia , Inibidores de Proteases/farmacologia , Animais , Antivirais/farmacologia , Infecções Bacterianas/tratamento farmacológico , Desenho de Fármacos , Protease de HIV/química , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Humanos , Serina Proteases , Proteínas não Estruturais Virais/antagonistas & inibidores
14.
J Pediatr Gastroenterol Nutr ; 66(5): 816-821, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29135822

RESUMO

OBJECTIVES: Whether premature infants have lower gastric protein digestive capacity than term infants and the extent to which human milk proteases contribute to overall gastric digestion are unknown and were investigated in this study. METHODS: Human milk and infant gastric samples were collected from 16 preterm (24-32 wk gestational age) and 6 term (38-40 wk gestational age) mother-infant pairs within a range of 5 to 42 days postnatal age. For each pair, an aliquot of human milk was adjusted to pH 4.5 and incubated for 2 hours at 37 °C to simulate the gastric conditions without pepsin (milkinc). Their gastric protein digestion capacity was measured as proteolysis (free N-terminals) and protease activities. Two-way analysis of variance followed by Tukey post hoc test was applied to compare measurements between preterm and term infants as well as among human milk, milkinc, and gastric samples. RESULTS: Measurements of gastric protein digestion were significantly lower in preterm infants than term infants. Overall milk protease activity did not differ between human milk samples from term- and preterm-delivering mothers. As protease activity did not increase with simulated gastric incubation, milk proteases likely contributed minimally to gastric digestion. CONCLUSIONS: Preterm infants have lower gastric protein digestion capacity than term infants, which could impair nutrient acquisition. Human milk proteases contribute minimally to overall gastric digestion. The limited activity of milk proteases suggests that these enzymes cannot compensate for the premature infant's overall lower gastric protein digestion.


Assuntos
Digestão/fisiologia , Recém-Nascido Prematuro/fisiologia , Proteínas do Leite/metabolismo , Leite Humano/metabolismo , Estômago/fisiologia , Adulto , Fluorometria , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Peptídeo Hidrolases/fisiologia , Proteólise , Espectrofotometria
15.
Biochim Biophys Acta Bioenerg ; 1858(8): 633-640, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28017650

RESUMO

The mitochondrial role in carcinogenesis and cancer progression is an area of active research, with many unresolved questions. Various aspects of altered mitochondrial function have been implicated in tumorigenesis and tumor progression, including mitochondrial dysfunction, a metabolic switch to aerobic glycolysis, and dysregulation of mitophagy. Mitophagy is a highly specific quality control process which eliminates dysfunctional mitochondria and promotes mitochondrial turnover, and is involved in the adaptation to nutrient stress by controlling mitochondrial mass. The dysregulation of mitochondrial turnover has both a positive and negative role in cancer. This review will begin with a basic overview of the molecular mechanisms of mitophagy, and highlight recent trends in mitophagy from cancer studies. We will conclude this review by discussing areas of research in normal mitophagy that have yet to be explored in the context of cancer such as mitochondrial proteases, the mitochondrial unfolded protein response, and mitokine action. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Assuntos
Transformação Celular Neoplásica , Mitofagia , Neoplasias/metabolismo , Animais , Proteínas de Caenorhabditis elegans/fisiologia , Progressão da Doença , Humanos , Dinâmica Mitocondrial , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/fisiologia , Mitofagia/fisiologia , Modelos Biológicos , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Neoplasias/etiologia , Neoplasias/patologia , Peptídeo Hidrolases/fisiologia , Isoformas de Proteínas/fisiologia , Resposta a Proteínas não Dobradas
16.
Exp Eye Res ; 163: 53-57, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28284957

RESUMO

The function of the meibomian gland in the upper and lower eyelids is critical to maintaining homeostasis at the ocular surface. Highly specialized meibocytes within the gland must differentiate and accumulate intracellular lipid droplets that are released into the tear film following rupture of the cell membrane. Proteases and their inhibitors have been recognized as key players in remodeling extracellular matrices and promoting the normal integrity of glandular tissue. They modulate a wide range of biological processes, such as cell proliferation and differentiation, and can contribute to disease when aberrantly expressed. Deciphering the role of proteolytic activity in the meibomian gland offers an opportunity to gain a more comprehensive and fundamental understanding of the developmental, physiological, and pathological processes associated with this gland.


Assuntos
Síndromes do Olho Seco/fisiopatologia , Glândulas Tarsais/fisiologia , Peptídeo Hidrolases/fisiologia , Proteólise , Envelhecimento/patologia , Envelhecimento/fisiologia , Matriz Extracelular/metabolismo , Humanos , Glândulas Tarsais/química , Glândulas Tarsais/patologia , Metaloproteases/metabolismo , Glândulas Sebáceas/metabolismo
18.
Gut ; 65(7): 1215-24, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27196587

RESUMO

The GI tract is the most exposed organ to proteases, both in physiological and pathophysiological conditions. For digestive purposes, the lumen of the upper GI tract contains large amounts of pancreatic proteases, but studies have also demonstrated increased proteolytic activity into mucosal tissues (both in the upper and lower GI tract), associated with pathological conditions. This review aims at outlining the evidences for dysregulated proteolytic homeostasis in GI diseases and the pathogenic mechanisms of increased proteolytic activity. The therapeutic potential of protease inhibition in GI diseases is discussed, with a particular focus on IBDs, functional GI disorders and colorectal cancer.


Assuntos
Gastroenteropatias/tratamento farmacológico , Gastroenteropatias/enzimologia , Peptídeo Hidrolases/fisiologia , Inibidores de Proteases/uso terapêutico , Animais , Doença Celíaca/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico
19.
Biochim Biophys Acta ; 1847(9): 910-4, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25528366

RESUMO

Progress in the field of regulated intramembrane proteolysis (RIP) in recent years has not surpassed plant biology. Nevertheless, reports on RIP in plants, and especially in chloroplasts, are still scarce. Of the four different families of intramembrane proteases, only two have been linked to chloroplasts so far, rhomboids and site-2 proteases (S2Ps). The lack of chloroplast-located rhomboid proteases was associated with reduced fertility and aberrations in flower morphology, probably due to perturbations in jasmonic acid biosynthesis, which occurs in chloroplasts. Mutations in homologues of S2P resulted in chlorophyll deficiency and impaired chloroplast development, through a yet unknown mechanism. To date, the only known substrate of RIP in chloroplasts is a PHD transcription factor, located in the envelope. Upon proteolytic cleavage by an unknown protease, the soluble N-terminal domain of this protein is released from the membrane and relocates to the nucleus, where it activates the transcription of the ABA response gene ABI4. Continuing studies on these proteases and substrates, as well as identification of the genes responsible for different chloroplast mutant phenotypes, are expected to shed more light on the roles of intramembrane proteases in chloroplast biology.


Assuntos
Proteínas de Membrana/metabolismo , Plastídeos/metabolismo , Peptídeo Hidrolases/fisiologia , Proteólise
20.
Biochem Biophys Res Commun ; 477(3): 388-94, 2016 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-27329811

RESUMO

The gene coding cereblon (CRBN) was originally identified in genetic linkage analysis of mild autosomal recessive nonsyndromic intellectual disability. CRBN has broad localization in both the cytoplasm and nucleus. However, the significance of nuclear CRBN remains unknown. In the present study, we aimed to elucidate the role of CRBN in the nucleus. First, we generated a series of CRBN deletion mutants and determined the regions responsible for the nuclear localization. Only CRBN protein lacking the N-terminal region was localized outside of the nucleus, suggesting that the N-terminal region is important for its nuclear localization. CRBN was also identified as a thalidomide-binding protein and component of the cullin-4-containing E3 ubiquitin ligase complex. Thalidomide has been reported to be involved in the regulation of the transcription factor Ikaros by CRBN-mediated degradation. To investigate the nuclear functions of CRBN, we performed co-immunoprecipitation experiments and evaluated the binding of CRBN to Ikaros. As a result, we found that CRBN was associated with Ikaros protein, and the N-terminal region of CRBN was required for Ikaros binding. In luciferase reporter gene experiments, CRBN modulated transcriptional activity of Ikaros. Furthermore, we found that CRBN modulated Ikaros-mediated transcriptional repression of the proenkephalin gene by binding to its promoter region. These results suggest that CRBN binds to Ikaros via its N-terminal region and regulates transcriptional activities of Ikaros and its downstream target, enkephalin.


Assuntos
Encefalinas/metabolismo , Fator de Transcrição Ikaros/genética , Neuroblastoma/metabolismo , Peptídeo Hidrolases/fisiologia , Transcrição Gênica/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Linhagem Celular Tumoral , Humanos , Neuroblastoma/patologia , Regiões Promotoras Genéticas , Ubiquitina-Proteína Ligases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA