Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 248
Filtrar
1.
Biomolecules ; 14(4)2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38672489

RESUMO

Colorectal cancer (CRC) is a leading cause of death worldwide. Conventional therapies are available with varying effectiveness. Acetate, a short-chain fatty acid produced by human intestinal bacteria, triggers mitochondria-mediated apoptosis preferentially in CRC but not in normal colonocytes, which has spurred an interest in its use for CRC prevention/therapy. We previously uncovered that acetate-induced mitochondrial-mediated apoptosis in CRC cells is significantly enhanced by the inhibition of the lysosomal protease cathepsin D (CatD), which indicates both mitochondria and the lysosome are involved in the regulation of acetate-induced apoptosis. Herein, we sought to determine whether mitochondrial function affects CatD apoptotic function. We found that enhancement of acetate-induced apoptosis by CatD inhibition depends on oligomycin A-sensitive respiration. Mechanistically, the potentiating effect is associated with an increase in cellular and mitochondrial superoxide anion accumulation and mitochondrial mass. Our results provide novel clues into the regulation of CatD function and the effect of tumor heterogeneity in the outcome of combined treatment using acetate and CatD inhibitors.


Assuntos
Apoptose , Catepsina D , Neoplasias Colorretais , Mitocôndrias , Oligomicinas , Humanos , Acetatos/farmacologia , Apoptose/efeitos dos fármacos , Catepsina D/metabolismo , Catepsina D/antagonistas & inibidores , Linhagem Celular Tumoral , Respiração Celular/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Neoplasias Colorretais/tratamento farmacológico , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Oligomicinas/farmacologia
2.
Bioorg Med Chem ; 57: 116646, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35121401

RESUMO

Cathepsin D (Cath D) has been evidenced as a potential target for cancer therapy. Our previous studies revealed that TB-9, a tasiamide B derivative, exhibited highly potent inhibition against Cath D with satisfactory selectivity over Cath E and BACE1. But this compound was inactive on cell level possibly due to poor membrane permeability. Herein, we report the design, synthesis, and evaluation of two novel Cath D inhibitors (2 and 3) which combining tasiamide B scaffold with a cell penetrating peptide (CPP) specifically targeting the endolysosomal compartment. The results revealed that 2 and 3 not only retained highly potent inhibition against Cath D, but also were active against MDA-MB-231 cell lines.


Assuntos
Catepsina D/antagonistas & inibidores , Oligopeptídeos/farmacologia , Inibidores de Proteases/farmacologia , Catepsina D/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Oligopeptídeos/síntese química , Oligopeptídeos/química , Inibidores de Proteases/síntese química , Inibidores de Proteases/química , Relação Estrutura-Atividade
3.
Cell Death Dis ; 13(2): 115, 2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-35121737

RESUMO

Cathepsin D (Cat D) is well known for its roles in metastasis, angiogenesis, proliferation, and carcinogenesis in cancer. Despite Cat D being a promising target in cancer cells, effects and underlying mechanism of its inhibition remain unclear. Here, we investigated the plausibility of using Cat D inhibition as an adjuvant or sensitizer for enhancing anticancer drug-induced apoptosis. Inhibition of Cat D markedly enhanced anticancer drug-induced apoptosis in human carcinoma cell lines and xenograft models. The inhibition destabilized Bcl-xL through upregulation of the expression of RNF183, an E3 ligase of Bcl-xL, via NF-κB activation. Furthermore, Cat D inhibition increased the proteasome activity, which is another important factor in the degradation of proteins. Cat D inhibition resulted in p62-dependent activation of Nrf2, which increased the expression of proteasome subunits (PSMA5 and PSMB5), and thereby, the proteasome activity. Overall, Cat D inhibition sensitized cancer cells to anticancer drugs through the destabilization of Bcl-xL. Furthermore, human renal clear carcinoma (RCC) tissues revealed a positive correlation between Cat D and Bcl-xL expression, whereas RNF183 and Bcl-xL expression indicated inverse correlation. Our results suggest that inhibition of Cat D is promising as an adjuvant or sensitizer for enhancing anticancer drug-induced apoptosis in cancer cells.


Assuntos
Antineoplásicos , Carcinoma de Células Renais , Catepsina D , Neoplasias Renais , Ubiquitina-Proteína Ligases , Antineoplásicos/farmacologia , Apoptose , Carcinoma de Células Renais/tratamento farmacológico , Catepsina D/antagonistas & inibidores , Linhagem Celular Tumoral , Humanos , Neoplasias Renais/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteína bcl-X/metabolismo
4.
Front Immunol ; 12: 675535, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335574

RESUMO

Background & Aims: The lysosomal enzyme, cathepsin D (CTSD) has been implicated in the pathogenesis of non-alcoholic steatohepatitis (NASH), a disease characterised by hepatic steatosis and inflammation. We have previously demonstrated that specific inhibition of the extracellular CTSD leads to improved metabolic features in Sprague-Dawley rats with steatosis. However, the individual roles of extracellular and intracellular CTSD in NASH are not yet known. In the current study, we evaluated the underlying mechanisms of extracellular and intracellular CTSD fractions in NASH-related metabolic inflammation using specific small-molecule inhibitors. Methods: Low-density lipoprotein receptor knock out (Ldlr-/-) mice were fed a high-fat, high cholesterol (HFC) diet for ten weeks to induce NASH. Further, to investigate the effects of CTSD inhibition, mice were injected either with an intracellular (GA-12) or extracellular (CTD-002) CTSD inhibitor or vehicle control at doses of 50 mg/kg body weight subcutaneously once in two days for ten weeks. Results: Ldlr-/- mice treated with extracellular CTSD inhibitor showed reduced hepatic lipid accumulation and an associated increase in faecal bile acid levels as compared to intracellular CTSD inhibitor-treated mice. Furthermore, in contrast to intracellular CTSD inhibition, extracellular CTSD inhibition switched the systemic immune status of the mice to an anti-inflammatory profile. In line, label-free mass spectrometry-based proteomics revealed that extra- and intracellular CTSD fractions modulate proteins belonging to distinct metabolic pathways. Conclusion: We have provided clinically translatable evidence that extracellular CTSD inhibition shows some beneficial metabolic and systemic inflammatory effects which are distinct from intracellular CTSD inhibition. Considering that intracellular CTSD inhibition is involved in essential physiological processes, specific inhibitors capable of blocking extracellular CTSD activity, can be promising and safe NASH drugs.


Assuntos
Catepsina D/fisiologia , Inflamação/etiologia , Metabolismo dos Lipídeos , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Animais , Ácidos e Sais Biliares/análise , Catepsina D/antagonistas & inibidores , Feminino , Inflamação/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Proteômica , Receptores de LDL/fisiologia
5.
Bioorg Med Chem ; 29: 115879, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33271453

RESUMO

Cathepsin D, an aspartyl protease, is an attractive therapeutic target for various diseases, primarily cancer and osteoarthritis. However, despite several small molecule cathepsin D inhibitors being developed, that are highly potent, most of them show poor microsomal stability, which in turn limits their clinical translation. Herein, we describe the design, optimization and evaluation of a series of novel non-peptidic acylguanidine based small molecule inhibitors of cathepsin D. Optimization of our hit compound 1a (IC50 = 29 nM) led to the highly potent mono sulphonamide analogue 4b (IC50 = 4 nM), however with poor microsomal stability (HLM: 177 and MLM: 177 µl/min/mg). To further improve the microsomal stability while retaining the potency, we carried out an extensive structure-activity relationship screen which led to the identification of our optimised lead 24e (IC50 = 45 nM), with an improved microsomal stability (HLM: 59.1 and MLM: 86.8 µl/min/mg). Our efforts reveal that 24e could be a good starting point or potential candidate for further preclinical studies against diseases where Cathepsin D plays an important role.


Assuntos
Catepsina D/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Catepsina D/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Microssomos Hepáticos/química , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
6.
Biomolecules ; 10(11)2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33182454

RESUMO

Finding effective neuroprotective strategies to combat various neurodegenerative disorders still remain a clinically unmet need. Methyl caffeate (MC), a naturally occurring ester of caffeic acid, possesses antioxidant and anti-inflammatory activities; however, its role in neuroprotection is less investigated. In order to better characterize neuroprotective properties of MC, we tested its effectiveness in various models of neuronal cell injury in human neuroblastoma SH-SY5Y cells and in mouse primary neuronal cell cultures. MC at micromolar concentrations attenuated neuronal cell damage induced by hydrogen peroxide (H2O2) in undifferentiated and neuronal differentiated SH-SY5Y cells as well as in primary cortical neurons. This effect was associated with inhibition of both caspase-3 and cathepsin D but without involvement of the PI3-K/Akt pathway. MC was neuroprotective when given before and during but not after the induction of cell damage by H2O2. Moreover, MC was protective against 6-OHDA-evoked neurotoxicity in neuronal differentiated SH-SY5Y cells via inhibition of necrotic and apoptotic processes. On the other hand, MC was ineffective in models of excitotoxicity (induced by glutamate or oxygen-glucose deprivation) and even moderately augmented cytotoxic effects of the classical apoptotic inducer, staurosporine. Finally, in undifferentiated neuroblastoma cells MC at higher concentrations (above 50 microM) induced cell death and when combined with the chemotherapeutic agent, doxorubicin, it increased the cell damaging effects of the latter compound. Thus, neuroprotective properties of MC appear to be limited to certain models of neurotoxicity and depend on its concentrations and time of administration.


Assuntos
Ácidos Cafeicos/farmacologia , Caspase 3/metabolismo , Inibidores de Caspase/farmacologia , Catepsina D/antagonistas & inibidores , Peróxido de Hidrogênio/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Inibidores de Proteases/farmacologia
7.
Adv Exp Med Biol ; 1259: 1-16, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32578168

RESUMO

Cancer remains a major and leading health problem worldwide. Lack of early diagnosis, chemoresistance, and recurrence of cancer means vast research and development are required in this area. The complexity of the tumor microenvironment in the biological milieu poses greater challenges in having safer, selective, and targeted therapies. Existing strategies such as chemotherapy, radiotherapy, and antiangiogenic therapies moderately improve progression-free survival; however, they come with side effects that reduce quality of life. Thus, targeting potential candidates in the microenvironment, such as extracellular cathepsin D (CathD) which has been known to play major pro-tumorigenic roles in breast and ovarian cancers, could be a breakthrough in cancer treatment, specially using novel treatment modalities such as immunotherapy and nanotechnology-based therapy. This chapter discusses CathD as a pro-cancerous, more specifically a proangiogenic factor, that acts bi-functionally in the tumor microenvironment, and possible ways of targeting the protein therapeutically.


Assuntos
Neoplasias da Mama/patologia , Catepsina D , Neoplasias Ovarianas/patologia , Microambiente Tumoral , Animais , Neoplasias da Mama/tratamento farmacológico , Catepsina D/antagonistas & inibidores , Feminino , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Qualidade de Vida , Microambiente Tumoral/efeitos dos fármacos
8.
Biochem Biophys Res Commun ; 527(1): 238-241, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32446374

RESUMO

Aspartic proteases are important biomarkers of human disease and interesting targets for modulation of immune response via MHC class II antigen processing inhibition. The lack of inhibitors with sufficient selectivity hampers precise analysis of the role of cathepsin E and napsin A in samples containing the ubiquitous and highly abundant homolog cathepsin D. Grassystatins from marine cyanobacteria show promising selectivity for cathepsin E but contain several ester bonds that make their synthesis cumbersome and thus limit availability of the inhibitors. Herewith, we present grassystatin-derived cathepsin E inhibitors with greatly facilitated synthesis but retained selectivity profile. We demonstrate their affinity and selectivity with both enzyme kinetic assays and streptavidin-based pull-down from cells and mouse organs. Our findings suggest that grassystatin-like inhibitors are useful tools for targeted inhibition of cathepsin E and thus provide a novel approach for cancer and immunology research.


Assuntos
Catepsina D/antagonistas & inibidores , Catepsina E/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Peptídeos/farmacologia , Catepsina D/metabolismo , Catepsina E/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Células HEK293 , Humanos , Estrutura Molecular , Peptídeos/síntese química , Peptídeos/química , Proteínas Recombinantes/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade
9.
Mol Carcinog ; 59(6): 651-660, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32253787

RESUMO

Postoperative radiotherapy combined with chemotherapy is a commonly used treatment for glioblastoma (GBM) but radiotherapy often fails to achieve the expected results mainly due to tumor radioresistance. In this study, we established a radioresistant subline from human glioma cell line U251 and found that Cathepsin D (CTSD), a gene closely related to the clinical malignancy and prognosis in glioma, had higher expression level in radioresistant clones than that in parental cells, and knocking down CTSD by small interfering RNA (siRNA) or its inhibitor Pepstatin-A increased the radiosensitivity. The level of autophagy was enhanced in the radioresistant GBM cells compared with its parent cells, and silencing autophagy by light chain 3 (LC3) siRNA significantly sensitized GBM cells to ionizing radiation (IR). Moreover, the protein expression level of CTSD was positively correlated with the autophagy marker LC3 II/I and negatively correlated with P62 after IR in radioresistant cells. As expected, through the combination of Western blot and immunofluorescence assays, inhibition of CTSD increased the formation of autophagosomes, while decreased the formation of autolysosomes, which indicating an attenuated autophagy level, leading to radiosensitization ultimately. Our results revealed for the first time that CTSD regulated the radiosensitivity of glioblastoma by affecting the fusion of autophagosomes and lysosomes. In significance, CTSD might be a potential molecular biomarker and a new therapeutic target in glioblastoma.


Assuntos
Autofagia , Neoplasias Encefálicas/radioterapia , Catepsina D/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Glioblastoma/radioterapia , RNA Interferente Pequeno/genética , Tolerância a Radiação/genética , Apoptose , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Catepsina D/genética , Proliferação de Células , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Prognóstico , Radiação Ionizante , Taxa de Sobrevida , Células Tumorais Cultivadas
10.
J Med Chem ; 63(4): 1576-1596, 2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-32003991

RESUMO

Human cathepsin D (CatD), a pepsin-family aspartic protease, plays an important role in tumor progression and metastasis. Here, we report the development of biomimetic inhibitors of CatD as novel tools for regulation of this therapeutic target. We designed a macrocyclic scaffold to mimic the spatial conformation of the minimal pseudo-dipeptide binding motif of pepstatin A, a microbial oligopeptide inhibitor, in the CatD active site. A library of more than 30 macrocyclic peptidomimetic inhibitors was employed for scaffold optimization, mapping of subsite interactions, and profiling of inhibitor selectivity. Furthermore, we solved high-resolution crystal structures of three macrocyclic inhibitors with low nanomolar or subnanomolar potency in complex with CatD and determined their binding mode using quantum chemical calculations. The study provides a new structural template and functional profile that can be exploited for design of potential chemotherapeutics that specifically inhibit CatD and related aspartic proteases.


Assuntos
Catepsina D/antagonistas & inibidores , Catepsina D/metabolismo , Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , Sítios de Ligação , Materiais Biomiméticos/síntese química , Materiais Biomiméticos/química , Materiais Biomiméticos/metabolismo , Materiais Biomiméticos/toxicidade , Células CACO-2 , Catepsina D/química , Ensaios Enzimáticos , Humanos , Cinética , Estrutura Molecular , Pepstatinas/química , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/toxicidade , Inibidores de Proteases/síntese química , Inibidores de Proteases/toxicidade , Ligação Proteica , Relação Estrutura-Atividade
11.
Neurotox Res ; 37(3): 525-542, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31960265

RESUMO

Necroptosis, a recently discovered form of non-apoptotic programmed cell death, can be implicated in many pathological conditions including neuronal cell death. Moreover, an inhibition of this process by necrostatin-1 (Nec-1) has been shown to be neuroprotective in in vitro and in vivo models of cerebral ischemia. However, the involvement of this type of cell death in oxidative stress-induced neuronal cell damage is less recognized. Therefore, we tested the effects of Nec-1, an inhibitor of necroptosis, in the model of hydrogen peroxide (H2O2)-induced cell damage in human neuroblastoma SH-SY5Y and murine hippocampal HT-22 cell lines. The data showed that Nec-1 (10-40 µM) attenuated the cell death induced by H2O2 in undifferentiated (UN-) and neuronal differentiated (RA-) SH-SY5Y cells with a higher efficacy in the former cell type. Moreover, Nec-1 partially reduced cell damage induced by 6-hydroxydopamine in UN- and RA-SH-SY5Y cells. The protective effect of Nec-1 was of similar magnitude as the effect of a caspase-3 inhibitor in both cell phenotypes and this effect were not potentiated after combined treatment. Furthermore, the non-specific apoptosis and necroptosis inhibitor curcumin augmented the beneficial effect of Nec-1 against H2O2-evoked cell damage albeit only in RA-SH-SY5Y cells. Next, it was found that the mechanisms of neuroprotective effect of Nec-1 against H2O2-induced cell damage in SH-SY5Y cells involved the inhibition of lysosomal protease, cathepsin D, but not caspase-3 or calpain activities. In HT-22 cells, Nec-1 was protective in two models of oxidative stress (H2O2 and glutamate) and that effect was blocked by a caspase inhibitor. Our data showed neuroprotective effects of the necroptosis inhibitor, Nec-1, against oxidative stress-induced cell damage and pointed to involvement of cathepsin D inhibition in the mechanism of its action. Moreover, a cell type-specific interplay between necroptosis and apoptosis has been demonstrated.


Assuntos
Catepsina D/antagonistas & inibidores , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Imidazóis/administração & dosagem , Indóis/administração & dosagem , Necroptose/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Animais , Inibidores de Caspase/administração & dosagem , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Curcumina/administração & dosagem , Humanos , Peróxido de Hidrogênio/administração & dosagem , Camundongos
12.
Biochem Biophys Res Commun ; 523(1): 239-245, 2020 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-31862139

RESUMO

Hyperglycemia is an independent risk factor for diabetic heart failure. However, the mechanisms that mediate hyperglycemia-induced cardiac damage remain poorly understood. Previous studies have shown an association between lysosomal dysfunction and diabetic heart injury. The present study examined if mimicking hyperglycemia in cultured cardiomyocytes could induce lysosomal membrane permeabilization (LMP), leading to the release of lysosome enzymes and subsequent cell death. High glucose (HG) reduced the number of lysosomes with acidic pH as shown by a fluorescent pH indicator. Also, HG induced lysosomal membrane injury as shown by an accumulation of Galectin3-RFP puncta, which was accompanied by the leakage of cathepsin D (CTSD), an aspartic protease that normally resides within the lysosomal lumen. Furthermore, CTSD expression was increased in HG-cultured cardiomyocytes and in the hearts of 2 mouse models of type 1 diabetes. Either CTSD knockdown with siRNA or inhibition of CTSD activity by pepstatin A markedly diminished HG-induced cardiomyocyte death, while CTSD overexpression exaggerated HG-induced cell death. Together, these results suggested that HG increased CTSD expression, induced LMP and triggered CTSD release from the lysosomes, which collectively contributed to HG-induced cardiomyocyte injury.


Assuntos
Catepsina D/metabolismo , Morte Celular , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Lisossomos/metabolismo , Lisossomos/patologia , Miócitos Cardíacos/metabolismo , Animais , Catepsina D/antagonistas & inibidores , Catepsina D/genética , Células Cultivadas , Humanos , Concentração de Íons de Hidrogênio , Microscopia de Fluorescência , Miócitos Cardíacos/patologia , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley
13.
Molecules ; 24(19)2019 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-31547147

RESUMO

The first total syntheses of izenamides A, B, and C, which are depsipeptides inhibitor of cathepsin D, were accomplished. In addition, the stereochemistry of izenamide B was confirmed by our syntheses. The key features of our synthetic route involve the avoidance of critical 2,5-diketopiperazine (DKP) formation and the minimization of epimerization during the coupling of amino acids for the target peptides.


Assuntos
Catepsina D/antagonistas & inibidores , Depsipeptídeos/síntese química , Depsipeptídeos/farmacologia , Depsipeptídeos/química , Dicetopiperazinas/química
14.
Biomolecules ; 9(5)2019 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-31060228

RESUMO

Dietary and lifestyle changes are leading to an increased occurrence of non-alcoholic fatty liver disease (NAFLD). Using a hyperlipidemic murine model for non-alcoholic steatohepatitis (NASH), we have previously demonstrated that the lysosomal protease cathepsin D (CTSD) is involved with lipid dysregulation and inflammation. However, despite identifying CTSD as a major player in NAFLD pathogenesis, the specific role of extracellular CTSD in NAFLD has not yet been investigated. Given that inhibition of intracellular CTSD is highly unfavorable due to its fundamental physiological function, we here investigated the impact of a highly specific and potent small-molecule inhibitor of extracellular CTSD (CTD-002) in the context of NAFLD. Treatment of bone marrow-derived macrophages with CTD-002, and incubation of hepatic HepG2 cells with a conditioned medium derived from CTD-002-treated macrophages, resulted in reduced levels of inflammation and improved cholesterol metabolism. Treatment with CTD-002 improved hepatic steatosis in high fat diet-fed rats. Additionally, plasma levels of insulin and hepatic transaminases were significantly reduced upon CTD-002 administration. Collectively, our findings demonstrate for the first time that modulation of extracellular CTSD can serve as a novel therapeutic modality for NAFLD.


Assuntos
Catepsina D/antagonistas & inibidores , Espaço Extracelular/enzimologia , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Inibidores de Proteases/uso terapêutico , Alanina Transaminase/metabolismo , Animais , Aspartato Aminotransferases/metabolismo , Catepsina D/metabolismo , Células Cultivadas , Dieta Hiperlipídica , Células Hep G2 , Humanos , Inflamação/patologia , Lipoproteínas LDL , Fígado/efeitos dos fármacos , Fígado/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Inibidores de Proteases/farmacologia , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
15.
Bioconjug Chem ; 30(7): 1979-1985, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30990716

RESUMO

Protease inhibitors are used as both research tools and therapeutics. Many of these inhibitors consist of substrate amino acid sequence-derived structure with a transition state mimic to interact with the active site of the protease, suppressing enzymatic activity. However, once they bind, macrodilution or protein denaturation is required to remove them, limiting their usage. In this study, we describe a removable protease inhibitor, which is a directly biotinylated analogue to control the activities of HIV-1 protease and human cathepsin D. In the substrate cleavage assay, we observed that the nanomolar inhibitory activities were lost upon the addition of streptavidin, while the enzymatic activities sufficiently recovered. HIV-1 protease mixed with the removable inhibitor, avoiding autolysis, was still active to be detected by adding streptavidin after one year at room temperature. We also observed that the inhibitor was an effective eluent for the simple detection of the activity of proteases purified from human serum and cells. These results demonstrate that direct biotinylation of protease inhibitors could be a novel method for controlling the enzymatic activity from OFF to ON. We proposed the phenomenon that binding equilibrium of inhibitor was shifted from protease to streptavidin with higher affinity, named "inhibitor stripping action by affinity competition", or ISAAC. We anticipate that ISAAC could be applicable for preservatives of proteases and activity-based diagnosis of protease related diseases. Furthermore, removable inhibitor to be designed for targeted proteases changing the inhibitor structure may elucidate enzymatic activity in intrinsic form with natural modifications from various biological samples.


Assuntos
Inibidores de Proteases/isolamento & purificação , Biotinilação , Catepsina D/antagonistas & inibidores , Desenho de Fármacos , Protease de HIV/química , Protease de HIV/metabolismo , HIV-1/enzimologia , Humanos , Modelos Moleculares , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia
16.
Int J Mol Sci ; 20(6)2019 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-30884823

RESUMO

Triple-negative breast cancers (TNBCs) are more aggressive than other breast cancer (BC) subtypes and lack effective therapeutic options. Unraveling marker events of TNBCs may provide new directions for development of strategies for targeted TNBC therapy. Herein, we reported that Annexin A1 (AnxA1) and Cathepsin D (CatD) are highly expressed in MDA-MB-231 (TNBC lineage), compared to MCF-10A and MCF-7. Since the proposed concept was that CatD has protumorigenic activity associated with its ability to cleave AnxA1 (generating a 35.5 KDa fragment), we investigated this mechanism more deeply using the inhibitor of CatD, Pepstatin A (PepA). Fourier Transform Infrared (FTIR) spectroscopy demonstrated that PepA inhibits CatD activity by occupying its active site; the OH bond from PepA interacts with a CO bond from carboxylic acids of CatD catalytic aspartate dyad, favoring the deprotonation of Asp33 and consequently inhibiting CatD. Treatment of MDA-MB-231 cells with PepA induced apoptosis and autophagy processes while reducing the proliferation, invasion, and migration. Finally, in silico molecular docking demonstrated that the catalytic inhibition comprises Asp231 protonated and Asp33 deprotonated, proving all functional results obtained. Our findings elucidated critical CatD activity in TNBC cell trough AnxA1 cleavage, indicating the inhibition of CatD as a possible strategy for TNBC treatment.


Assuntos
Anexina A1/genética , Catepsina D/genética , Simulação de Acoplamento Molecular , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Domínio Catalítico/efeitos dos fármacos , Catepsina D/antagonistas & inibidores , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Pepstatinas/farmacologia , Espectroscopia de Infravermelho com Transformada de Fourier , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
17.
J Pept Sci ; 25(4): e3154, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30734395

RESUMO

Cathepsin D (Cath D) is overexpressed and hypersecreted by malignant tumors and involved in the progress of tumor invasion, proliferation, metastasis, and apoptosis. Cath D has been considered as a potential target to treat cancer. Our previous studies revealed that tasiamide B derivatives TB-9 and TB-11 exhibited high potent inhibition against Cath D and other aspartic proteases, but their molecular weights are still high, and the role of each residue is unknown yet. Based on this, two series of tasiamide B derivatives have been designed, synthesized, and evaluated for their inhibitory activity against Cath D/Cath E/BACE1. Enzymatic assays revealed that the target compound 1 with lower molecule weight showed good inhibitory activity against Cath D with IC50 of 3.29 nM and satisfactory selectivity over Cath E (72-fold) and BACE1 (295-fold), which could be a valuable template for the design of highly potent and selective Cath D inhibitors.


Assuntos
Catepsina D/antagonistas & inibidores , Desenho de Fármacos , Oligopeptídeos/farmacologia , Inibidores de Proteases/farmacologia , Catepsina D/metabolismo , Relação Dose-Resposta a Droga , Estrutura Molecular , Oligopeptídeos/síntese química , Oligopeptídeos/química , Inibidores de Proteases/síntese química , Inibidores de Proteases/química , Relação Estrutura-Atividade
18.
J Immunother Cancer ; 7(1): 29, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30717773

RESUMO

BACKGROUND: Triple-negative breast cancer (TNBC) treatment is currently restricted to chemotherapy. Hence, tumor-specific molecular targets and/or alternative therapeutic strategies for TNBC are urgently needed. Immunotherapy is emerging as an exciting treatment option for TNBC patients. The aspartic protease cathepsin D (cath-D), a marker of poor prognosis in breast cancer (BC), is overproduced and hypersecreted by human BC cells. This study explores whether cath-D is a tumor cell-associated extracellular biomarker and a potent target for antibody-based therapy in TNBC. METHODS: Cath-D prognostic value and localization was evaluated by transcriptomics, proteomics and immunohistochemistry in TNBC. First-in-class anti-cath-D human scFv fragments binding to both human and mouse cath-D were generated using phage display and cloned in the human IgG1 λ format (F1 and E2). Anti-cath-D antibody biodistribution, antitumor efficacy and in vivo underlying mechanisms were investigated in TNBC MDA-MB-231 tumor xenografts in nude mice. Antitumor effect was further assessed in TNBC patient-derived xenografts (PDXs). RESULTS: High CTSD mRNA levels correlated with shorter recurrence-free survival in TNBC, and extracellular cath-D was detected in the tumor microenvironment, but not in matched normal breast stroma. Anti-cath-D F1 and E2 antibodies accumulated in TNBC MDA-MB-231 tumor xenografts, inhibited tumor growth and improved mice survival without apparent toxicity. The Fc function of F1, the best antibody candidate, was essential for maximal tumor inhibition in the MDA-MB-231 model. Mechanistically, F1 antitumor response was triggered through natural killer cell activation via IL-15 upregulation, associated with granzyme B and perforin production, and the release of antitumor IFNγ cytokine. The F1 antibody also prevented the tumor recruitment of immunosuppressive tumor-associated macrophages M2 and myeloid-derived suppressor cells, a specific effect associated with a less immunosuppressive tumor microenvironment highlighted by TGFß decrease. Finally, the antibody F1 inhibited tumor growth of two TNBC PDXs, isolated from patients resistant or not to neo-adjuvant chemotherapy. CONCLUSION: Cath-D is a tumor-specific extracellular target in TNBC suitable for antibody-based therapy. Immunomodulatory antibody-based strategy against cath-D is a promising immunotherapy to treat patients with TNBC.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Catepsina D/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Anticorpos Monoclonais/farmacocinética , Antineoplásicos Imunológicos/farmacocinética , Catepsina D/genética , Catepsina D/imunologia , Linhagem Celular Tumoral , Feminino , Humanos , Imunoterapia , Camundongos Nus , RNA Mensageiro/metabolismo , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
19.
J Cell Biochem ; 120(6): 10662-10669, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30652348

RESUMO

Earlier studies showed that the oxidant menadione (MD) induces apoptosis in certain cells and also has anticancer effects. Most of these studies emphasized the role of the mitochondria in this process. However, the engagement of other organelles is less known. Particularly, the role of lysosomes and their proteolytic system, which participates in apoptotic cell death, is still unclear. The aim of this study was to investigate the role of lysosomal cathepsins on molecular signaling in MD-induced apoptosis in U937 cells. MD treatment induced translocation of cysteine cathepsins B, C, and S, and aspartic cathepsin D. Once in the cytosol, some cathepsins cleaved the proapoptotic molecule, Bid, in a process that was completely prevented by E64d, a general inhibitor of cysteine cathepsins, and partially prevented by the pancaspase inhibitor, z-VAD-fmk. Upon loss of the mitochondrial membrane potential, apoptosome activation led to caspase-9 processing, activation of caspase-3-like caspases, and poly (ADP-ribose) polymerase cleavage. Notably, the endogenous protein inhibitor, stefin B, was degraded by cathepsin D and caspases. This process was prevented by z-VAD-fmk, and partially by pepstatin A-penetratin. These findings suggest that the cleaved Bid protein acts as an amplifier of apoptotic signaling through mitochondria, thus enhancing the activity of cysteine cathepsins following stefin B degradation.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Cistatina B/genética , Regulação Neoplásica da Expressão Gênica , Lisossomos/efeitos dos fármacos , Vitamina K 3/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Apoptose/genética , Apoptossomas/efeitos dos fármacos , Apoptossomas/metabolismo , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Catepsina B/antagonistas & inibidores , Catepsina B/genética , Catepsina B/metabolismo , Catepsina C/antagonistas & inibidores , Catepsina C/genética , Catepsina C/metabolismo , Catepsina D/antagonistas & inibidores , Catepsina D/genética , Catepsina D/metabolismo , Catepsinas/antagonistas & inibidores , Catepsinas/genética , Catepsinas/metabolismo , Cistatina B/metabolismo , Humanos , Leucina/análogos & derivados , Leucina/farmacologia , Lisossomos/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Pepstatinas/farmacologia , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Inibidores de Proteases/farmacologia , Proteólise/efeitos dos fármacos , Transdução de Sinais , Células U937
20.
Eur J Med Chem ; 163: 344-352, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30529637

RESUMO

Following up the open initiative of anti-malarial drug discovery, a GlaxoSmithKline (GSK) phenotypic screening hit was developed to generate hydroxyethylamine based plasmepsin (Plm) inhibitors exhibiting growth inhibition of the malaria parasite Plasmodium falciparum at nanomolar concentrations. Lead optimization studies were performed with the aim of improving Plm inhibition selectivity versus the related human aspartic protease cathepsin D (Cat D). Optimization studies were performed using Plm IV as a readily accessible model protein, the inhibition of which correlates with anti-malarial activity. Guided by sequence alignment of Plms and Cat D, selectivity-inducing structural motifs were modified in the S3 and S4 sub-pocket occupying substituents of the hydroxyethylamine inhibitors. This resulted in potent anti-malarials with an up to 50-fold Plm IV/Cat D selectivity factor. More detailed investigation of the mechanism of action of the selected compounds revealed that they inhibit maturation of the P. falciparum subtilisin-like protease SUB1, and also inhibit parasite egress from erythrocytes. Our results indicate that the anti-malarial activity of the compounds is linked to inhibition of the SUB1 maturase plasmepsin subtype Plm X.


Assuntos
Antimaláricos/farmacologia , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Catepsina D/antagonistas & inibidores , Peptidomiméticos/farmacologia , Animais , Antimaláricos/química , Ácido Aspártico Endopeptidases/genética , Catepsina D/genética , Eritrócitos/parasitologia , Etilaminas/antagonistas & inibidores , Humanos , Peptidomiméticos/uso terapêutico , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Inibidores de Proteases/química , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...