Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 367
Filtrar
1.
BMC Cancer ; 24(1): 445, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38600469

RESUMO

BACKGROUND: Proprotein convertase subtilisin/kexin type 9 (PCSK9), the last member of the proprotein convertase family, functions as a classic regulator of low-density lipoprotein (LDL) by interacting with low-density lipoprotein receptor (LDLR). Recent studies have shown that PCSK9 can affect the occurrence and development of tumors and can be used as a novel therapeutic target. However, a comprehensive pan-cancer analysis of PCSK9 has yet to be conducted. METHODS: The potential oncogenic effects of PCSK9 in 33 types of tumors were explored based on the datasets of The Cancer Genome Atlas (TCGA) dataset. In addition, the immune regulatory role of PCSK9 inhibition was evaluated via in vitro cell coculture and the tumor-bearing mouse model. Finally, the antitumor efficacy of targeted PCSK9 combined with OVA-II vaccines was verified. RESULTS: Our results indicated that PCSK9 was highly expressed in most tumor types and was significantly correlated with late disease stage and poor prognosis. Additionally, PCSK9 may regulate the tumor immune matrix score, immune cell infiltration, immune checkpoint expression, and major histocompatibility complex expression. Notably, we first found that dendritic cell (DC) infiltration and major histocompatibility complex-II (MHC-II) expression could be upregulated by PCSK9 inhibition and improve CD8+ T cell activation in the tumor immune microenvironment, thereby achieving potent tumor control. Combining PCSK9 inhibitors could enhance the efficacies of OVA-II tumor vaccine monotherapy. CONCLUSIONS: Conclusively, our pan-cancer analysis provided a more comprehensive understanding of the oncogenic and immunoregulatory roles of PCSK9 and demonstrated that targeting PCSK9 could increase the efficacy of long peptide vaccines by upregulating DC infiltration and MHC-II expression on the surface of tumor cells. This study reveals the critical oncogenic and immunoregulatory roles of PCSK9 in various tumors and shows the promise of PCSK9 as a potent immunotherapy target.


Assuntos
Genes MHC da Classe II , Imunoterapia , Neoplasias , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Animais , Camundongos , Antígenos de Histocompatibilidade , Lipoproteínas LDL , Neoplasias/genética , Neoplasias/terapia , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertases/antagonistas & inibidores , Receptores de LDL/genética , Microambiente Tumoral
2.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34349020

RESUMO

Family with sequence similarity 20C (Fam20C), the major protein kinase in the secretory pathway, generates the vast majority of the secreted phosphoproteome. However, the regulatory mechanisms of Fam20C transport, secretion, and function remain largely unexplored. Here, we show that Fam20C exists as a type II transmembrane protein within the secretory compartments, with its N-terminal signal peptide-like region serving as a membrane anchor for Golgi retention. The secretion and kinase activity of Fam20C are governed by site-1 protease (S1P), a key regulator of cholesterol homeostasis. We find that only mature Fam20C processed by S1P functions in osteoblast differentiation and mineralization. Together, our findings reveal a unique mechanism for Fam20C secretion and activation via proteolytic regulation, providing a molecular link between biomineralization and lipid metabolism.


Assuntos
Caseína Quinase I/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/metabolismo , Motivos de Aminoácidos , Animais , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Caseína Quinase I/genética , Diferenciação Celular/efeitos dos fármacos , Proteínas da Matriz Extracelular/genética , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Camundongos , Mutação , Osteoblastos/citologia , Osteoblastos/metabolismo , Pró-Proteína Convertases/antagonistas & inibidores , Pró-Proteína Convertases/genética , Domínios Proteicos , Transporte Proteico , Pirrolidinas/farmacologia , Via Secretória , Serina Endopeptidases/genética
3.
Cell Death Dis ; 12(8): 717, 2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-34285190

RESUMO

Renal cell carcinoma (RCC) cells have increased lipogenesis and cholesterol synthesis. Sterol regulatory element-binding protein-1 (SREBP1) is cleaved by site 1 protease (S1P) to release the transcriptionally active amino-terminal domain. PF-429242 is a potent and competitive S1P inhibitor. We here tested its activity in RCC cells. In established and primary human RCC cells, PF-429242 potently inhibited cell proliferation, migration, and invasion. The S1P inhibitor provoked apoptosis activation in RCC cells. Furthermore, shRNA-mediated S1P silencing or CRISPR/Cas9-induced S1P knockout led to RCC cell growth inhibition and apoptosis activation. Conversely, ectopic overexpression of SREBP1 or S1P augmented RCC cell proliferation and migration. Daily i.v. injection of a single dose of PF-429242 robustly inhibited RCC xenograft growth in severe combined immunodeficiency mice. Additionally, intratumoral injection of S1P shRNA lentivirus inhibited RCC xenograft growth in mice. SREBP1, S1P, and its target gene low density lipoprotein receptor (LDLR) were significantly elevated in human RCC tissues. These results suggest that targeting S1P by PF-429242 inhibited RCC cell growth in vitro and in vivo.


Assuntos
Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Pró-Proteína Convertases/antagonistas & inibidores , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Inativação Gênica/efeitos dos fármacos , Humanos , Túbulos Renais/patologia , Masculino , Pessoa de Meia-Idade , Pró-Proteína Convertases/metabolismo , Pirrolidinas , Serina Endopeptidases/metabolismo , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Biochem Biophys Res Commun ; 558: 1-7, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33894672

RESUMO

ATF6 has two isoforms, ATF6α and ATF6ß, which are ubiquitously expressed type II transmembrane glycoproteins in the endoplasmic reticulum (ER). While the regulatory mechanisms and transcriptional roles of ATF6α in response to ER stress have been well-studied, those of its paralogue ATF6ß are less understood. Moreover, there is no specific cell-based reporter assay to monitor ATF6ß activation. Here, we developed a new cell-based reporter system that can monitor activation of endogenous ATF6ß. This system expresses a chimeric protein containing a synthetic transcription factor followed by the transmembrane domain and C-terminal luminal domain of ATF6ß. Under ER stress conditions, the chimeric protein was cleaved by regulated intramembrane proteolysis (RIP) to liberate the N-terminal synthetic transcription factor, which induced luciferase expression in the HeLa Luciferase Reporter cell line. This new stable reporter cell line will be an innovative tool to investigate RIP of ATF6ß.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Fator 6 Ativador da Transcrição/química , Fator 6 Ativador da Transcrição/genética , Linhagem Celular , Ditiotreitol/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/genética , Genes Reporter , Humanos , Luciferases de Vaga-Lume/química , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Pró-Proteína Convertases/antagonistas & inibidores , Domínios Proteicos , Pirrolidinas/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Serina Endopeptidases , Transativadores/química , Transativadores/genética , Transativadores/metabolismo
5.
Hypertension ; 77(2): 405-416, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33280408

RESUMO

Activation of PRR ([pro]renin receptor) contributes to enhancement of intrarenal RAS and renal medullary α-ENaC and thus elevated blood pressure during Ang II (angiotensin II) infusion. The goal of the present study was to test whether such action of PRR was mediated by sPRR (soluble PRR), generated by S1P (site-1 protease), a newly identified PRR cleavage protease. F1 B6129SF1/J mice were infused for 6 days with control or Ang II at 300 ng/kg per day alone or in combination with S1P inhibitor PF-429242 (PF), and blood pressure was monitored by radiotelemetry. S1P inhibition significantly attenuated Ang II-induced hypertension accompanied with suppressed urinary and renal medullary renin levels and expression of renal medullary but not renal cortical α-ENaC expression. The effects of S1P inhibition were all reversed by supplement with histidine-tagged sPRR termed as sPRR-His. Ussing chamber technique was performed to determine amiloride-sensitive short-circuit current, an index of ENaC activity in confluent mouse cortical collecting duct cell line cells exposed for 24 hours to Ang II, Ang II + PF, or Ang II + PF + sPRR-His. Ang II-induced ENaC activity was blocked by PF, which was reversed by sPRR-His. Together, these results support that S1P-derived sPRR mediates Ang II-induced hypertension through enhancement of intrarenal renin level and activation of ENaC.


Assuntos
Angiotensina II/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/genética , Pró-Proteína Convertases/antagonistas & inibidores , Pirrolidinas/farmacologia , Receptores de Superfície Celular/genética , Animais , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Camundongos , Receptores de Superfície Celular/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Serina Endopeptidases , Receptor de Pró-Renina
6.
Biochim Biophys Acta Mol Cell Res ; 1868(3): 118912, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33249002

RESUMO

Despite continuous exertion made, colon cancer still represents a major health problem and its incidence continues being high worldwide. There is growing evidence in support of the cancer stem cells (CSCs) being central in the initiation of this cancer, and CSCs have been the focus of various studies for the identification of new ways of treatment. Lately, the proprotein convertases (PCs) were reported to regulate the maturation and expression of various molecules involved in the malignant phenotype of colon cancer cells, however, the identity of the molecules regulated by these serine proteases in CSCs is unknown. In this study, we used the general PCs inhibitor, the Decanoyl-RVKR-chloromethylketone (Decanoyl-RVKR-CMK) that inhibits all the PCs found in the secretory pathway, and analyzed its effect on CSCs using RNA-seq analysis. Remarkably, from the only 9 up-regulated genes in the human SW620-derived sphere-forming cells, we identified 7 of the 11 human metallothioneins, all of them localized on chromosome 16, and zinc related proteins as downstream effectors of the PCs. The importance of these molecules in the regulation of cell proliferation, differentiation and chemoresistance, and their reported potential tumor suppressor role and loss in colon cancer patients associated with worse prognosis, suggests that targeting PCs in the control of the malignant phenotype of CSCs is a new potential therapeutic strategy in colon cancer.


Assuntos
Clorometilcetonas de Aminoácidos/farmacologia , Neoplasias do Colo/enzimologia , Perfilação da Expressão Gênica/métodos , Metalotioneína/genética , Células-Tronco Neoplásicas/enzimologia , Regulação para Cima , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Células-Tronco Neoplásicas/química , Células-Tronco Neoplásicas/efeitos dos fármacos , Pró-Proteína Convertases/antagonistas & inibidores , Análise de Sequência de RNA , Sequenciamento do Exoma
7.
Acta Derm Venereol ; 100(10): adv00157, 2020 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-32449780

RESUMO

Although recent therapeutic developments raise hope, melanoma remains a devastating disease with a need for new treatment targets. In other tumours prohormone convertases have been shown to be pro-tumourigenic as they are involved in processing preforms of matrix-metalloproteinases, growth factors and adhesion molecules. The aim of this study was to look for new treatment options for melanoma, by investigating the role of the prohormone convertase Paired basic Amino acid-Cleaving Enzyme 4 (PACE4/PCSK6) in melanoma cell lines and human melanoma tissue. PACE4-transfected A375 melanoma cells displayed significantly increased proliferation, MMP-2 production, gelatinase activity and migratory capacity in vitro compared with sham-transfected cells. In vivo, elevated PACE4 expression resulted in significantly increased tumour growth on immunodeficient mice. In the majority of 45 human primary melanomas and melanoma metastases ex vivo PACE4 immunoreactivity was detectable, while it was absent in in situ melanomas. These results indicate PACE4 as a regulator of melanoma cell aggressiveness.


Assuntos
Melanoma/enzimologia , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/metabolismo , Neoplasias Cutâneas/enzimologia , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/patologia , Camundongos Pelados , Camundongos SCID , Terapia de Alvo Molecular , Invasividade Neoplásica , Pró-Proteína Convertases/antagonistas & inibidores , Pró-Proteína Convertases/genética , Serina Endopeptidases/genética , Inibidores de Serino Proteinase/uso terapêutico , Transdução de Sinais , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Carga Tumoral
8.
Hipertens. riesgo vasc ; 37(1): 42-44, ene.-mar. 2020.
Artigo em Espanhol | IBECS | ID: ibc-188673

RESUMO

Planteamos un paciente de 62 años que precisa tratamiento hipolipemiante con inhibidor de la proproteína convertasa subtilisina/kexina tipo9 (PCSK9) en el que empíricamente decidimos cambiar un fármaco a otro de los dos disponibles en la actualidad, obteniendo diferente respuesta. Nuestro objetivo es transmitir nuestra experiencia y reflexionar sobre una posible opción terapéutica en los pacientes en los que excepcionalmente esto pudiera ocurrir


The case is presented of a 62-year-old patient who required lipid-lowering therapy with proprotein convertase subtilisin/kexin type9 (PCSK9). It was empirically decided to change one drug to another of the two currently available, obtaining a different response. Our objective is to present our experience and to consider a possible therapeutic option in patients in whom, exceptionally, this could happen


Assuntos
Humanos , Masculino , Pessoa de Meia-Idade , Pró-Proteína Convertase 9/administração & dosagem , Pró-Proteína Convertases/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Hipercolesterolemia/tratamento farmacológico , Pressão Arterial/efeitos dos fármacos , Pró-Proteína Convertases/metabolismo , Índice de Massa Corporal , Lipoproteínas LDL/efeitos dos fármacos
9.
Cancer Lett ; 473: 50-61, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-31899298

RESUMO

Many cancers occur from locations of inflammation due to chronic irritation and/or infection. Tumor microenvironment contains various different inflammatory cells and mediators that orchestrate diverse neoplastic processes, including proliferation, survival, adhesion and migration. In parallel, tumor cells have adapted some of the signaling molecules used by inflammatory cells, such as selectins and chemokines as well as their receptors for invasion, extravasation and subsequently metastasis. Expression and/or activation of the majority of these molecules is mediated by the proprotein convertases (PCs); proteases expressed by both tumor cells and inflammatory cells. This review analyzes the potential role of these enzymatic system in inflammation-associated cancer impacting on the malignant and metastatic potential of cancer cells, describing the possible use of PCs as a new anti-inflammatory therapeutic approach to tumor progression and metastasis.


Assuntos
Carcinogênese/imunologia , Inflamação/tratamento farmacológico , Metástase Neoplásica/imunologia , Neoplasias/imunologia , Pró-Proteína Convertases/metabolismo , Animais , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Carcinogênese/efeitos dos fármacos , Quimiocinas/imunologia , Quimiocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Inflamação/imunologia , Inflamação/patologia , Metástase Neoplásica/prevenção & controle , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Pró-Proteína Convertases/antagonistas & inibidores , Selectinas/imunologia , Selectinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
10.
J Virol ; 94(5)2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-31826992

RESUMO

Antibody-dependent enhancement (ADE) of viral entry has been a major concern for epidemiology, vaccine development, and antibody-based drug therapy. However, the molecular mechanism behind ADE is still elusive. Coronavirus spike protein mediates viral entry into cells by first binding to a receptor on the host cell surface and then fusing viral and host membranes. In this study, we investigated how a neutralizing monoclonal antibody (MAb), which targets the receptor-binding domain (RBD) of Middle East respiratory syndrome (MERS) coronavirus spike, mediates viral entry using pseudovirus entry and biochemical assays. Our results showed that MAb binds to the virus surface spike, allowing it to undergo conformational changes and become prone to proteolytic activation. Meanwhile, MAb binds to cell surface IgG Fc receptor, guiding viral entry through canonical viral-receptor-dependent pathways. Our data suggest that the antibody/Fc-receptor complex functionally mimics viral receptor in mediating viral entry. Moreover, we characterized MAb dosages in viral-receptor-dependent, Fc-receptor-dependent, and both-receptors-dependent viral entry pathways, delineating guidelines on MAb usages in treating viral infections. Our study reveals a novel molecular mechanism for antibody-enhanced viral entry and can guide future vaccination and antiviral strategies.IMPORTANCE Antibody-dependent enhancement (ADE) of viral entry has been observed for many viruses. It was shown that antibodies target one serotype of viruses but only subneutralize another, leading to ADE of the latter viruses. Here we identify a novel mechanism for ADE: a neutralizing antibody binds to the surface spike protein of coronaviruses like a viral receptor, triggers a conformational change of the spike, and mediates viral entry into IgG Fc receptor-expressing cells through canonical viral-receptor-dependent pathways. We further evaluated how antibody dosages impacted viral entry into cells expressing viral receptor, Fc receptor, or both receptors. This study reveals complex roles of antibodies in viral entry and can guide future vaccine design and antibody-based drug therapy.


Assuntos
Anticorpos Antivirais/imunologia , Anticorpos Facilitadores , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Internalização do Vírus , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Linhagem Celular , Dipeptidil Peptidase 4/metabolismo , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , Peptídeo Hidrolases/metabolismo , Pró-Proteína Convertases/antagonistas & inibidores , Pró-Proteína Convertases/metabolismo , Conformação Proteica , Domínios Proteicos , Multimerização Proteica , Receptores Fc/metabolismo , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Receptores Virais/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Tripsina/metabolismo
11.
Cells ; 8(12)2019 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-31766635

RESUMO

During tumorigenesis, macrophages are recruited by tumors and orientated towards a pro-tumoral phenotype. One of the main anti-tumoral immunotherapy consists of their re-polarization in an anti-tumoral phenotype. We have demonstrated that the inhibition of proprotein convertase 1/3 combined with TLR4 activation in macrophages is a promising strategy. These macrophages display pro-inflammatory and anti-tumoral phenotypes. A hallmark is a stronger activation of the pro-inflammatory NFKB pathway. We believe that this can be explained by a modification of TLR4 expression at the cell surface or MYD88 cleavage since it exhibits a potential cleavage site for proprotein convertases. We tested these hypotheses through immunofluorescence and Western blot experiments. A proteomics study was also performed to test the sensitivity of these macrophages to IL-10. We demonstrated that these macrophages treated with LPS showed a quicker re-expression of TLR4 at the cell surface. The level of MYD88 was also higher when TLR4 was internalized. Moreover, these macrophages were resistant to the pro-tumoral effect of IL-10 and still produced pro-inflammatory factors. This established that the sensitivity to anti-inflammatory molecules and the length of TLR4 desensitization were reduced in these macrophages. Therefore, during antitumoral immunotherapy, a repeated stimulation of TLR4 may reactivate PC1/3 inhibited macrophages even in an anti-inflammatory environment.


Assuntos
Interleucina-10/metabolismo , Macrófagos/metabolismo , Pró-Proteína Convertases/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Lipopolissacarídeos/farmacologia , Fenótipo , Pró-Proteína Convertases/antagonistas & inibidores , Pró-Proteína Convertases/deficiência , Ratos
12.
FEBS J ; 286(22): 4597-4620, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31276291

RESUMO

The basic proprotein convertases (PCs) furin, PC1/3, PC2, PC5/6, PACE4, PC4, and PC7 are promising drug targets for human diseases. However, developing selective inhibitors remains challenging due to overlapping substrate recognition motifs and limited structural information. Classical drug screening approaches for basic PC inhibitors involve homogeneous biochemical assays using soluble recombinant enzymes combined with fluorogenic substrate peptides that may not accurately recapitulate the complex cellular context of the basic PC-substrate interaction. Herein we report basic PC sensor (BPCS), a novel cell-based molecular sensor that allows rapid screening of candidate inhibitors and their selectivity toward individual basic PCs within mammalian cells. BPCS consists of Gaussia luciferase linked to a sortilin-1 membrane anchor via a cleavage motif that allows efficient release of luciferase specifically if individual basic PCs are provided in the same membrane. Screening of selected candidate peptidomimetic inhibitors revealed that BPCS can readily distinguish between general and selective PC inhibitors in a high-throughput screening format. The robust and cost-effective assay format of BPCS makes it suitable to identify novel specific small-molecule inhibitors against basic PCs for therapeutic application. Its cell-based nature will allow screening for drug targets in addition to the catalytically active mature enzyme, including maturation, transport, and cellular factors that modulate the enzyme's activity. This broadened 'target range' will enhance the likelihood to identify novel small-molecule compounds that inhibit basic PCs in a direct or indirect manner and represents a conceptual advantage.


Assuntos
Técnicas Biossensoriais/métodos , Descoberta de Drogas/métodos , Inibidores Enzimáticos/farmacologia , Peptidomiméticos/farmacologia , Pró-Proteína Convertases/metabolismo , Células A549 , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Técnicas Biossensoriais/normas , Descoberta de Drogas/normas , Inibidores Enzimáticos/química , Genes Reporter , Células HEK293 , Células HeLa , Ensaios de Triagem em Larga Escala/métodos , Ensaios de Triagem em Larga Escala/normas , Humanos , Luciferases/genética , Luciferases/metabolismo , Peptidomiméticos/química , Pró-Proteína Convertases/antagonistas & inibidores , Sensibilidade e Especificidade
15.
JCI Insight ; 4(7)2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30944256

RESUMO

The antidiuretic hormone vasopressin (AVP), acting through its type 2 receptor (V2R) in the collecting duct (CD), critically controls urine concentrating capability. Here, we report that site-1 protease-derived (S1P-derived) soluble (pro)renin receptor (sPRR) participates in regulation of fluid homeostasis via targeting V2R. In cultured inner medullary collecting duct (IMCD) cells, AVP-induced V2R expression was blunted by a PRR antagonist, PRO20; a PRR-neutralizing antibody; or a S1P inhibitor, PF-429242. In parallel, sPRR release was increased by AVP and reduced by PF-429242. Administration of histidine-tagged sPRR, sPRR-His, stimulated V2R expression and also reversed the inhibitory effect of PF-429242 on the expression induced by AVP. PF-429242 treatment in C57/BL6 mice impaired urine concentrating capability, which was rescued by sPRR-His. This observation was recapitulated in mice with renal tubule-specific deletion of S1P. During the pharmacological or genetic manipulation of S1P alone or in combination with sPRR-His, the changes in urine concentration were paralleled with renal expression of V2R and aquaporin-2 (AQP2). Together, these results support that S1P-derived sPRR exerts a key role in determining renal V2R expression and, thus, urine concentrating capability.


Assuntos
Capacidade de Concentração Renal/fisiologia , Túbulos Renais Coletores/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Vasopressinas/metabolismo , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Aquaporina 2/genética , Células Cultivadas , Células Epiteliais , Capacidade de Concentração Renal/efeitos dos fármacos , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Fragmentos de Peptídeos/farmacologia , Cultura Primária de Células , Pró-Proteína Convertases/antagonistas & inibidores , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Pirrolidinas/farmacologia , Ratos , Receptores de Vasopressinas/genética , Renina/metabolismo , Renina/farmacologia , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Urotélio/citologia , ATPases Vacuolares Próton-Translocadoras
16.
Nat Commun ; 10(1): 285, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30655525

RESUMO

There are no approved therapies for Ebola virus infection. Here, to find potential therapeutic targets, we perform a screen for genes essential for Ebola virus (EBOV) infection. We identify GNPTAB, which encodes the α and ß subunits of N-acetylglucosamine-1-phosphate transferase. We show that EBOV infection of a GNPTAB knockout cell line is impaired, and that this is reversed by reconstituting GNPTAB expression. Fibroblasts from patients with mucolipidosis II, a disorder associated with mutations in GNPTAB, are refractory to EBOV, whereas cells from their healthy parents support infection. Impaired infection correlates with loss of the expression of cathepsin B, known to be essential for EBOV entry. GNPTAB activity is dependent upon proteolytic cleavage by the SKI-1/S1P protease. Inhibiting this protease with the small-molecule PF-429242 blocks EBOV entry and infection. Disruption of GNPTAB function may represent a strategy for a host-targeted therapy for EBOV.


Assuntos
Antivirais/farmacologia , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/tratamento farmacológico , Mucolipidoses/patologia , Transferases (Outros Grupos de Fosfato Substituídos)/genética , Internalização do Vírus/efeitos dos fármacos , Células A549 , Animais , Antivirais/uso terapêutico , Catepsina B/metabolismo , Chlorocebus aethiops , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Fibroblastos , Técnicas de Inativação de Genes , Doença pelo Vírus Ebola/virologia , Humanos , Mucolipidoses/genética , Pró-Proteína Convertases/antagonistas & inibidores , Pró-Proteína Convertases/metabolismo , Inibidores de Proteases/farmacologia , Pirrolidinas/farmacologia , Serina Endopeptidases/metabolismo , Transferases (Outros Grupos de Fosfato Substituídos)/antagonistas & inibidores , Células Vero , Sequenciamento Completo do Genoma
17.
ChemMedChem ; 14(6): 673-685, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30680958

RESUMO

The activation of viral glycoproteins by the host protease furin is an essential step in the replication of numerous pathogenic viruses. Thus, effective inhibitors of furin could serve as broad-spectrum antiviral drugs. A crystal structure of an inhibitory hexapeptide derivative in complex with furin served as template for the rational design of various types of new cyclic inhibitors. Most of the prepared derivatives are relatively potent furin inhibitors with inhibition constants in the low nanomolar or even sub-nanomolar range. For seven derivatives the crystal structures in complex with furin could be determined. In three complexes, electron density was found for the entire inhibitor. In the other cases the structures could be determined only for the P6/P5-P1 segments, which directly interact with furin. The cyclic derivatives together with two non-cyclic reference compounds were tested as inhibitors of the proteolytic activation and replication of respiratory syncytial virus in cells. Significant antiviral activity was found for both linear reference inhibitors, whereas a negligible efficacy was determined for the cyclic derivatives.


Assuntos
Inibidores Enzimáticos/farmacologia , Furina/antagonistas & inibidores , Compostos Macrocíclicos/farmacologia , Pró-Proteína Convertases/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Humanos , Compostos Macrocíclicos/síntese química
18.
Biochemistry ; 57(51): 6973-6983, 2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30499665

RESUMO

Overexpression of lysyl oxidase-like 2 (LOXL2) is associated with several hepatic and vascular fibrotic diseases and tumor progression in some aggressive cancers. Secreted LOXL2 promotes extracellular matrix cross-linking by catalyzing the oxidative deamination of peptidyl lysine. A great deal remains to be learned about the post-translational modifications of LOXL2, including whether such modifications modulate enzymatic and disease-promoting activities; such knowledge would inform the development of potential therapies. We discovered that upon secretion in cell culture, LOXL2 undergoes proteolytic processing of the first two of four scavenger receptor cysteine-rich domains at the N-terminus. A similar pattern of processing was also evident in tissue extracts from an invasive ductal carcinoma patient. Processing occurred at 314Arg-315Phe-316Arg-317Lys↓-318Ala-, implicating proprotein convertases. siRNA-mediated knockdown of proprotein convertases (furin, PACE4, and PC5/6), as well as incubation with their recombinant forms, showed that PACE4 is the major protease that acts on extracellular LOXL2. Unlike LOX, which requires cleavage of its propeptide for catalytic activation, cleavage of LOXL2 was not essential for tropoelastin oxidation or for cross-linking of collagen type IV in vitro. However, in the latter case, processing enhanced the extent of collagen cross-linking ∼2-fold at ≤10 nM LOXL2. These results demonstrate an important difference in the regulatory mechanisms for LOX and LOXL2 catalytic activity. Moreover, they pave the way for further studies of potential differential functions of LOXL2 isoforms in fibrosis and tumor progression.


Assuntos
Aminoácido Oxirredutases/metabolismo , Aminoácido Oxirredutases/química , Aminoácido Oxirredutases/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Neoplasias da Mama/enzimologia , Linhagem Celular , Colágeno Tipo IV/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Mutagênese Sítio-Dirigida , Pró-Proteína Convertases/antagonistas & inibidores , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Domínios Proteicos , Processamento de Proteína Pós-Traducional , Proteína-Lisina 6-Oxidase/química , Proteína-Lisina 6-Oxidase/genética , Proteína-Lisina 6-Oxidase/metabolismo , RNA Interferente Pequeno/genética , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
19.
J Med Chem ; 61(24): 11250-11260, 2018 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-30501188

RESUMO

Paired basic amino acid cleaving enzyme 4 (PACE4), a serine endoprotease of the proprotein convertases family, has been recognized as a promising target for prostate cancer. We previously reported a selective and potent peptide-based inhibitor for PACE4, named the multi-Leu peptide (Ac-LLLLRVKR-NH2 sequence), which was then modified into a more potent and stable compound named C23 with the following structure: Ac-dLeu-LLLRVK-Amba (Amba: 4-amidinobenzylamide). Despite improvements in both in vitro and in vivo profiles of C23, its selectivity for PACE4 over furin was significantly reduced. We examined other Arg-mimetics instead of Amba to regain the lost selectivity. Our results indicated that the replacement of Amba with 5-(aminomethyl)picolinimidamide increased affinity for PACE4 and restored selectivity. Our results also provide a better insight on how structural differences between S1 pockets of PACE4 and furin could be employed in the rational design of selective inhibitors.


Assuntos
Antineoplásicos/farmacologia , Pró-Proteína Convertases/antagonistas & inibidores , Inibidores de Serino Proteinase/química , Inibidores de Serino Proteinase/farmacologia , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desenho de Fármacos , Humanos , Masculino , Simulação de Acoplamento Molecular , Pró-Proteína Convertases/química , Pró-Proteína Convertases/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Inibidores de Serino Proteinase/metabolismo , Relação Estrutura-Atividade
20.
J Med Chem ; 61(18): 8457-8467, 2018 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-30180568

RESUMO

The serine protease, PACE4, is a proprotein convertase that plays a substantial role in malignancy of prostate cancer. Our initial selective PACE4 inhibitor (Ac-LLLLRVKR-NH2) has evolved to the current lead compound C23 (Ac-dLeu-LLLRVK-Amba), which is active both in vitro and in vivo. By screening natural residues, except Cys, in C-terminal P1' position, it was established that increasing hydrophobicity was improving cell permeability, which was directly translated into PCa cells antiproliferative activity. This cell antiproliferation enhancement seems independent from effect of P1' residue on PACE4 affinity. Replacement of P1-Amba of C23 by Acpa (( S)-2-amino-3-(4-carbamimidoylphenyl)propanoic acid) followed by addition of tryptamine in P1' resulted in compound 32 exhibiting superior PCa cells antiproliferative activity over the reference compound C23 (3-fold). This study sheds light on key factors that improve cell penetrating property and antiproliferative activity of PACE4 inhibitors.


Assuntos
Antineoplásicos/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Pró-Proteína Convertases/antagonistas & inibidores , Neoplasias da Próstata/patologia , Antineoplásicos/química , Inibidores Enzimáticos/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Masculino , Modelos Moleculares , Estrutura Molecular , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Conformação Proteica , Serina Endopeptidases , Relação Estrutura-Atividade , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...