Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.400
Filtrar
1.
J Photochem Photobiol B ; 255: 112919, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38677261

RESUMO

Endolysosomes perform a wide range of cellular functions, including nutrient sensing, macromolecule digestion and recycling, as well as plasma membrane repair. Because of their high activity in cancerous cells, endolysosomes are attractive targets for the development of novel cancer treatments. Light-activated compounds termed photosensitizers (PS) can catalyze the oxidation of specific biomolecules and intracellular organelles. To selectively damage endosomes and lysosomes, HT-29 colorectal cancer cells were incubated with nanomolar concentrations of meso-tetraphenylporphine disulfonate (TPPS2a), an amphiphilic PS taken up via endocytosis and activated by green light (522 nm, 2.1 J.cm-1). Several cellular responses were characterized by a combination of immunofluorescence and immunoblotting assays. We showed that TPPS2a photosensitization blocked autophagic flux without extensive endolysosomal membrane rupture. Nevertheless, there was a severe functional failure of endolysosomes due to a decrease in CTSD (cathepsin D, 55%) and CTSB (cathepsin B, 52%) maturation. PSAP (prosaposin) processing (into saposins) was also considerably impaired, a fact that could be detrimental to glycosphingolipid homeostasis. Therefore, photosensitization of HT-29 cells previously incubated with a low concentration of TPPS2a promotes endolysosomal dysfunction, an effect that can be used to improve cancer therapies.


Assuntos
Autofagia , Lisossomos , Fármacos Fotossensibilizantes , Humanos , Células HT29 , Lisossomos/metabolismo , Lisossomos/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Autofagia/efeitos da radiação , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Endossomos/metabolismo , Endossomos/efeitos dos fármacos , Catepsinas/metabolismo , Catepsinas/antagonistas & inibidores , Luz , Porfirinas/farmacologia , Porfirinas/química , Catepsina D/metabolismo , Catepsina B/metabolismo
2.
Inorg Chem ; 63(17): 7973-7983, 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38616353

RESUMO

Dysregulated cathepsin activity is linked to various human diseases including metabolic disorders, autoimmune conditions, and cancer. Given the overexpression of cathepsin in the tumor microenvironment, cathepsin inhibitors are promising pharmacological agents and drug delivery vehicles for cancer treatment. In this study, we describe the synthesis and photochemical and biological assessment of a dual-action agent based on ruthenium that is conjugated with a cathepsin inhibitor, designed for both photodynamic therapy (PDT) and photochemotherapy (PCT). The ruthenium-cathepsin inhibitor conjugate was synthesized through an oxime click reaction, combining a pan-cathepsin inhibitor based on E64d with the Ru(II) PCT/PDT fragment [Ru(dqpy)(dppn)], where dqpy = 2,6-di(quinoline-2-yl)pyridine and dppn = benzo[i]dipyrido[3,2-a:2',3'-c]phenazine. Photochemical investigations validated the conjugate's ability to release a triazole-containing cathepsin inhibitor for PCT and to generate singlet oxygen for PDT upon exposure to green light. Inhibition studies demonstrated the conjugate's potent and irreversible inactivation of purified and intracellular cysteine cathepsins. Two Ru(II) PCT/PDT agents based on the [Ru(dqpy)(dppn)] moiety were evaluated for photoinduced cytotoxicity in 4T1 murine triple-negative breast cancer cells, L929 fibroblasts, and M0, M1, and M2 macrophages. The cathepsin inhibitor conjugate displayed notable selectivity for inducing cell death under irradiation compared to dark conditions, mitigating toxicity in the dark observed with the triazole control complex [Ru(dqpy)(dppn)(MeTz)]2+ (MeTz = 1-methyl-1H-1,2,4-triazole). Notably, our lead complex is among a limited number of dual PCT/PDT agents activated with green light.


Assuntos
Catepsinas , Luz , Fotoquimioterapia , Fármacos Fotossensibilizantes , Rutênio , Humanos , Rutênio/química , Rutênio/farmacologia , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/síntese química , Catepsinas/antagonistas & inibidores , Catepsinas/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Estrutura Molecular , Linhagem Celular Tumoral , Complexos de Coordenação/farmacologia , Complexos de Coordenação/química , Complexos de Coordenação/síntese química , Animais , Camundongos , Sobrevivência Celular/efeitos dos fármacos , Luz Verde
3.
Eur J Pharmacol ; 919: 174826, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35157914

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the dysregulation of various cell types and immunological pathways. Autoantibodies play an important role in its pathogenesis. The presence of autoantibodies suggests that self-antigen presentation through major histocompatibility complex (MHC) class II on antigen presenting cells is involved in the pathogenesis of autoimmune diseases, including SLE. Cathepsin S (CatS) is a key protease for antigen peptide loading onto lysosomal/endosomal MHC class II molecules through invariant chain degradation to promote antigen presentation. Inhibition of CatS is therefore expected to suppress antigen presentation via MHC class II, T and B cell activation, and antibody production from B cells. Here, we report the pharmacological profile of ASP1617, a novel CatS inhibitor. ASP1617 induced invariant chain accumulation and decreased the expression level of MHC class ΙΙ on the cell surface in both mouse and human B cells. Further, ASP1617 prevented DO11.10 mice T cell proliferation to ovalbumin antigen. We investigated the effects of ASP1617 and mycophenolate mofetil (MMF) on the development of lupus-like nephritis in NZB/W F1 mice, a widely used SLE mouse model. Oral administration of ASP1617 suppressed anti-dsDNA IgG, prevented progression of lupus-like glomerulonephritis, and significantly prevented proteinuria excretion. In contrast, MMF did not suppress anti-dsDNA IgG. Further, we found that plasma and/or urine CatS levels were increased in specimens from NZB/W F1 mice and several SLE patients. These results indicate that CatS may be an attractive therapeutic target for the treatment of SLE.


Assuntos
Catepsinas/antagonistas & inibidores , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Inibidores de Proteases/farmacologia , Administração Oral , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos NZB , Inibidores de Proteases/administração & dosagem , Inibidores de Proteases/uso terapêutico
4.
Chem Biol Interact ; 353: 109811, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-35016848

RESUMO

Pancreatic cancer will be the second leading cause of cancer-related mortality worldwide due to its high rate of metastasis. Cathepsins (CATs) are effectors of invasive growth in various cancers. Currently, targeting CATs represents an attractive strategy for the treatment of highly metastatic cancers with high CATs activity, such as pancreatic cancer. To develop a stronger antimetastatic agent, ASPER-29, a novel inhibitor of CATs designed by using the asperphenamate derivative BBP as a lead compound, was synthesized, and its therapeutic potential in pancreatic cancer metastasis was investigated in this study. Molecular docking and enzyme inhibition assays proved that ASPER-29 can inhibit the activity of CAT-L and CAT-S by binding with these enzymes in classical action modes. Furthermore, ASPER-29 significantly inhibited the activity of CAT-L and CAT-S but had no effect on their expression in PANC-1 and BxPC-3 cells. The in vitro antimetastatic activities of ASPER-29 were examined by wound healing and Transwell chamber assays. We found that ASPER-29 inhibited the migration and invasion of PANC-1 and BxPC-3 cells in a concentration-dependent manner. Moreover, the in vivo antimetastatic effects of ASPER-29 were confirmed in a mouse xenotransplantation model. H&E staining and immunohistochemistry assays of Ki67 and CEACAM6 proved that ASPER-29 treatment significantly blocked the metastasis of pancreatic cancer cells to lung and liver tissues. Additionally, the activity of both CAT-L and CAT-S was markedly inhibited in the lung and liver tissues of ASPER-29-administered mice compared with the mice in the model group, suggesting that the metastasis-blocking effect of ASPER-29 should be mediated via inhibition of the activity of CAT-L and CAT-S in pancreatic cancer cells. Together, our results demonstrated that ASPER-29, as a novel inhibitor of CAT-L and CAT-S, possessed the evident ability to block the metastasis of pancreatic cancer cells.


Assuntos
Catepsina L/antagonistas & inibidores , Catepsinas/antagonistas & inibidores , Movimento Celular/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Animais , Antígenos CD/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sítios de Ligação , Catepsina L/metabolismo , Catepsinas/metabolismo , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/metabolismo , Humanos , Fígado/metabolismo , Fígado/patologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Nus , Simulação de Acoplamento Molecular , Metástase Neoplásica , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , Inibidores de Proteases/uso terapêutico , Transplante Heterólogo
5.
Cell Mol Life Sci ; 79(1): 34, 2022 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-34989869

RESUMO

New therapeutic targets that could improve current antitumor therapy and overcome cancer resistance are urgently needed. Promising candidates are lysosomal cysteine cathepsins, proteolytical enzymes involved in various critical steps during cancer progression. Among them, cathepsin X, which acts solely as a carboxypeptidase, has received much attention. Our results indicate that the triazole-based selective reversible inhibitor of cathepsin X named Z9 (1-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-2-((4-isopropyl-4H-1,2,4-triazol-3-yl)thio)ethan-1-one) significantly reduces tumor progression, both in vitro in cell-based functional assays and in vivo in two independent tumor mouse models: the FVB/PyMT transgenic and MMTV-PyMT orthotopic breast cancer mouse models. One of the mechanisms by which cathepsin X contributes to cancer progression is the compensation of cathepsin-B activity loss. Our results confirm that cathepsin-B inhibition is compensated by an increase in cathepsin X activity and protein levels. Furthermore, the simultaneous inhibition of both cathepsins B and X with potent, selective, reversible inhibitors exerted a synergistic effect in impairing processes of tumor progression in in vitro cell-based assays of tumor cell migration and spheroid growth. Taken together, our data demonstrate that Z9 impairs tumor progression both in vitro and in vivo and can be used in combination with other peptidase inhibitors as an innovative approach to overcome resistance to antipeptidase therapy.


Assuntos
Catepsina B/antagonistas & inibidores , Catepsinas/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Carga Tumoral/efeitos dos fármacos , Animais , Catepsina B/metabolismo , Catepsinas/genética , Catepsinas/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Inibidores Enzimáticos/química , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos Transgênicos , Invasividade Neoplásica , Infiltração de Neutrófilos/efeitos dos fármacos
6.
Biomed Pharmacother ; 145: 112245, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34772578

RESUMO

Cathepsin S (CTSS), a lysosomal protease, belongs to a family of cysteine cathepsin proteases that promote degradation of damaged proteins in the endolysosomal pathway. Aberrant CTSS expression and regulation are associated with the pathogenesis of several diseases, including lung diseases. CTSS overexpression causes a variety of pathological processes, including pulmonary fibrosis, with increased CTSS secretion and accelerated extracellular matrix remodeling. Compared to many other cysteine cathepsin family members, CTSS has unique features that it presents limited tissue expression and retains its enzymatic activity at a neutral pH, suggesting its decisive involvement in disease microenvironments. In this review, we investigated the role of CTSS in lung disease, exploring recent studies that have indicated that CTSS mediates fibrosis in unique ways, along with its structure, substrates, and distinct regulation. We also outlined examples of CTSS inhibitors in clinical and preclinical development and proposed CTSS as a potential therapeutic target for pulmonary fibrosis.


Assuntos
Catepsinas/antagonistas & inibidores , Fibrose Pulmonar/tratamento farmacológico , Animais , Catepsinas/metabolismo , Desenvolvimento de Medicamentos , Matriz Extracelular/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Terapia de Alvo Molecular , Fibrose Pulmonar/fisiopatologia
8.
Signal Transduct Target Ther ; 6(1): 379, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34744168

RESUMO

In recent years, accumulating evidence has elucidated the role of lysosomes in dynamically regulating cellular and organismal homeostasis. Lysosomal changes and dysfunction have been correlated with the development of numerous diseases. In this review, we interpreted the key biological functions of lysosomes in four areas: cellular metabolism, cell proliferation and differentiation, immunity, and cell death. More importantly, we actively sought to determine the characteristic changes and dysfunction of lysosomes in cells affected by these diseases, the causes of these changes and dysfunction, and their significance to the development and treatment of human disease. Furthermore, we outlined currently available targeting strategies: (1) targeting lysosomal acidification; (2) targeting lysosomal cathepsins; (3) targeting lysosomal membrane permeability and integrity; (4) targeting lysosomal calcium signaling; (5) targeting mTOR signaling; and (6) emerging potential targeting strategies. Moreover, we systematically summarized the corresponding drugs and their application in clinical trials. By integrating basic research with clinical findings, we discussed the current opportunities and challenges of targeting lysosomes in human disease.


Assuntos
Catepsinas/genética , Doenças por Armazenamento dos Lisossomos/tratamento farmacológico , Lisossomos/genética , Serina-Treonina Quinases TOR/genética , Sinalização do Cálcio/efeitos dos fármacos , Catepsinas/antagonistas & inibidores , Morte Celular/genética , Diferenciação Celular/genética , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/genética , Proliferação de Células/genética , Humanos , Doenças por Armazenamento dos Lisossomos/genética , Lisossomos/efeitos dos fármacos , Lisossomos/patologia , Serina-Treonina Quinases TOR/antagonistas & inibidores
9.
Bioorg Chem ; 115: 105213, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34364050

RESUMO

Cathepsins K and S are closely related papain-like cysteine peptidases and potential therapeutic targets for metabolic and inflammatory diseases such as osteoporosis and arthritis. Here we describe the reduction of a previously characterized succinimide (2,5-dioxopyrrolidine)-containing hyperbolic inhibitor of cathepsin K (methyl (RS)-N-[1-(4-methoxyphenyl)-2,5-dioxopyrrolidin-3-yl]glycinate), to obtain a better and more selective compound (compound 4a - methyl (2,5-dioxopyrrolidin-3-yl)glycinate), which acted as a hyperbolic mixed inhibitor/activator similar to already known allosteric effectors of cathepsin K. We then investigated the potential of the succinimide scaffold as inhibitors of cathepsins K and/or S and synthesized a library of such compounds by 1,4-addition of α-amino acid esters and related compounds to N-substituted maleimides. From the generated library, we identified the first small molecule hyperbolic inhibitors of cathepsin S (methyl ((R)-2,5-dioxopyrrolidin-3-yl)-l-threoninate (compound R-4c) and 3-{[(1S,2R,3'S)-2-hydroxycyclohexyl]amino}pyrrolidine-2,5-dione (compound (1S,2R,3'S-10)). The former acted via a similar mechanism to compound 4a, while the latter was a hyperbolic specific inhibitor of cathepsin S. Given the versatility of the scaffold, the identified compounds will be used as the basis for the development of high-affinity hyperbolic inhibitors of the individual peptidases and to explore the potential of hyperbolic inhibitors for the inhibition of cysteine cathepsins in in vitro models.


Assuntos
Catepsina K/antagonistas & inibidores , Catepsinas/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Succinimidas/farmacologia , Catepsina K/metabolismo , Catepsinas/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Cinética , Estrutura Molecular , Relação Estrutura-Atividade , Succinimidas/síntese química , Succinimidas/química
10.
Front Immunol ; 12: 726984, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34421929

RESUMO

The moment a very old bacterial pathogen met a young virus from the 80's defined the beginning of a tragic syndemic for humanity. Such is the case for the causative agent of tuberculosis and the human immunodeficiency virus (HIV). Syndemic is by definition a convergence of more than one disease resulting in magnification of their burden. Both pathogens work synergistically contributing to speed up the replication of each other. Mycobacterium tuberculosis (Mtb) and HIV infections are in the 21st century among the leaders of morbidity and mortality of humankind. There is an urgent need for development of new approaches for prevention, better diagnosis, and new therapies for both infections. Moreover, these approaches should consider Mtb and HIV as a co-infection, rather than just as separate problems, to prevent further aggravation of the HIV-TB syndemic. Both pathogens manipulate the host immune responses to establish chronic infections in intracellular niches of their host cells. This includes manipulation of host relevant antimicrobial proteases such as cathepsins or their endogenous inhibitors. Here we discuss recent understanding on how Mtb and HIV interact with cathepsins and their inhibitors in their multifactorial functions during the pathogenesis of both infections. Particularly we will address the role on pathogen transmission, during establishment of intracellular chronic niches and in granuloma clinical outcome and tuberculosis diagnosis. This area of research will open new avenues for the design of innovative therapies and diagnostic interventions so urgently needed to fight this threat to humanity.


Assuntos
Catepsinas/imunologia , Infecções por HIV/imunologia , Tuberculose/imunologia , Animais , Catepsinas/antagonistas & inibidores , Granuloma/imunologia , Infecções por HIV/diagnóstico , Infecções por HIV/transmissão , Interações Hospedeiro-Patógeno , Humanos , Imunidade nas Mucosas , Mucosa/imunologia , Mucosa/microbiologia , Tuberculose/diagnóstico , Tuberculose/transmissão
11.
Drug Resist Updat ; 57: 100769, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34217999

RESUMO

Multidrug resistance (MDR) remains a major obstacle towards curative treatment of cancer. Despite considerable progress in delineating the basis of intrinsic and acquired MDR, the underlying molecular mechanisms remain to be elucidated. Emerging evidences suggest that dysregulation in endolysosomal compartments is involved in mediating MDR through multiple mechanisms, such as alterations in endosomes, lysosomes and autophagosomes, that traffic and biodegrade the molecular cargo through macropinocytosis, autophagy and endocytosis. For example, altered lysosomal pH, in combination with transcription factor EB (TFEB)-mediated lysosomal biogenesis, increases the sequestration of hydrophobic anti-cancer drugs that are weak bases, thereby producing an insufficient and off-target accumulation of anti-cancer drugs in MDR cancer cells. Thus, the use of well-tolerated, alkalinizing compounds that selectively block Vacuolar H⁺-ATPase (V-ATPase) may be an important strategy to overcome MDR in cancer cells and increase chemotherapeutic efficacy. Other mechanisms of endolysosomal-mediated drug resistance include increases in the expression of lysosomal proteases and cathepsins that are involved in mediating carcinogenesis and chemoresistance. Therefore, blocking the trafficking and maturation of lysosomal proteases or direct inhibition of cathepsin activity in the cytosol may represent novel therapeutic modalities to overcome MDR. Furthermore, endolysosomal compartments involved in catabolic pathways, such as macropinocytosis and autophagy, are also shown to be involved in the development of MDR. Here, we review the role of endolysosomal trafficking in MDR development and discuss how targeting endolysosomal pathways could emerge as a new therapeutic strategy to overcome chemoresistance in cancer.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Endossomos/metabolismo , Lisossomos/metabolismo , Animais , Autofagia/efeitos dos fármacos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Transporte Biológico/efeitos dos fármacos , Catepsinas/antagonistas & inibidores , Linhagem Celular Tumoral , Humanos , Concentração de Íons de Hidrogênio , ATPases Vacuolares Próton-Translocadoras/metabolismo
12.
Mediators Inflamm ; 2021: 6682657, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33828414

RESUMO

BACKGROUND: Elevated levels of the cysteine protease cathepsin S (CatS) are associated with chronic mucoobstructive lung diseases such as cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD). We have previously demonstrated that prophylactic treatment with a CatS inhibitor from birth reduces inflammation, mucus plugging, and lung tissue damage in juvenile ß-epithelial Na+ channel-overexpressing transgenic (ßENaC-Tg) mice with chronic inflammatory mucoobstructive lung disease. In this study, we build upon this work to examine the effects of therapeutic intervention with a CatS inhibitor in adult ßENaC-Tg mice with established disease. METHODS: ßENaC-Tg mice and wild-type (WT) littermates were treated with a CatS inhibitor from 4 to 6 weeks of age, and CatS-/- ßENaC-Tg mice were analysed at 6 weeks of age. Bronchoalveolar lavage (BAL) fluid inflammatory cell counts were quantified, and lung tissue destruction and mucus obstruction were analysed histologically. RESULTS: At 6 weeks of age, ßENaC-Tg mice developed significant airway inflammation, lung tissue damage, and mucus plugging when compared to WT mice. CatS-/- ßENaC-Tg mice and ßENaC-Tg mice receiving inhibitor had significantly reduced airway mononuclear and polymorphonuclear (PMN) cell counts as well as mucus plugging. However, in contrast to CatS-/- ßENaC-Tg mice, therapeutic inhibition of CatS in ßENaC-Tg mice had no effect on established emphysema-like lung tissue damage. CONCLUSIONS: These results suggest that while early CatS targeting may be required to prevent the onset and progression of lung tissue damage, therapeutic CatS targeting effectively inhibited airway inflammation and mucus obstruction. These results indicate the important role CatS may play in the pathogenesis and progression of mucoobstructive lung disease.


Assuntos
Catepsinas/antagonistas & inibidores , Fibrose Cística , Canais Epiteliais de Sódio , Animais , Fibrose Cística/patologia , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Inflamação/patologia , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Muco
13.
Theranostics ; 11(10): 4672-4687, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754020

RESUMO

Rationale: Oxaliplatin-induced peripheral neuropathy (OIPN) is a common adverse effect that causes delayed treatment and poor prognosis among colorectal cancer (CRC) patients. However, its mechanism remains elusive, and no effective treatment is available. Methods: We employed a prospective cohort study of adult patients with pathologically confirmed stage III CRC receiving adjuvant chemotherapy with an oxaliplatin-based regimen for investigating OIPN. To further validate the clinical manifestations and identify a potential therapeutic strategy, animal models, and in vitro studies on the mechanism of OIPN were applied. Results: Our work found that (1) consistent with clinical findings, OIPN was observed in animal models. Targeting the enzymatic activity of cathepsin S (CTSS) by pharmacological blockade and gene deficiency strategy alleviates the manifestations of OIPN. (2) Oxaliplatin treatment increases CTSS expression by enhancing cytosol translocation of interferon response factor 1 (IRF1), which then facilitates STIM-dependent store-operated Ca2+ entry homeostasis. (3) The cytokine array demonstrated an increase in anti-inflammatory cytokines and suppression of proinflammatory cytokines in mice treated with RJW-58. (4) Mechanistically, inhibiting CTSS facilitated olfactory receptors transcription factor 1 release from P300/CBP binding, which enhanced binding to the interleukin-10 (IL-10) promoter region, driving IL-10 downstream signaling pathway. (5) Serum CTSS expression is increased in CRC patients with oxaliplatin-induced neurotoxicity. Conclusions: We highlighted the critical role of CTSS in OIPN, which provides a therapeutic strategy for the common adverse side effects of oxaliplatin.


Assuntos
Catepsinas/genética , Neurônios/metabolismo , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Catepsinas/antagonistas & inibidores , Catepsinas/efeitos dos fármacos , Quimioterapia Adjuvante , Estudos de Coortes , Neoplasias Colorretais/tratamento farmacológico , Citocinas/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos , Feminino , Fluoruracila/uso terapêutico , Gânglios Espinais , Humanos , Técnicas In Vitro , Leucovorina/uso terapêutico , Masculino , Camundongos , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/metabolismo , Terapia de Alvo Molecular , Condução Nervosa , Neurônios/efeitos dos fármacos , Compostos Organoplatínicos/uso terapêutico , Oxaliplatina/efeitos adversos , Oxaliplatina/farmacologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Estudos Prospectivos
14.
J Cell Physiol ; 236(2): 1309-1320, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32657442

RESUMO

Myocardial ischemia/reperfusion (I/R) injury leads to high mortality and morbidity due to the incomplete understanding of the underlying mechanism and the consequent lack of effective therapy. The present study revealed and validated key candidate genes in relation to inflammation and apoptosis pathways underlying myocardial I/R injury. Cathepsin S was identified as the top hub protein based on the protein-protein interaction analysis, and, thus, its role during myocardial I/R injury was further investigated. Myocardial I/R in mice resulted in significantly increased levels of myocardial injury biomarkers (cardiac troponin I, lactic dehydrogenase, and creatinine kinase-MB) and inflammatory cytokines (interleukin-1ß [IL-1ß], IL-6, and tumor necrosis factor-α), elevated apoptosis rate, and upregulated protein expression of cleaved caspase-8, cleaved caspase-3, and cleaved poly ADP-ribose polymerase. These abovementioned changes were blocked by two different selective cathepsin S inhibitors, LY3000328 or MIV-247. Moreover, Kaplan-Meier survival plot showed that cathepsin S inhibition improved 21-day survival rate following myocardial I/R injury. This study demonstrated that the inhibition of cathepsin S alleviated myocardial I/R-induced injury by suppressing inflammation and apoptosis, which may be used in clinical applications of cardioprotection.


Assuntos
Benzopiranos/farmacologia , Carbamatos/farmacologia , Catepsinas/genética , Infarto do Miocárdio/tratamento farmacológico , Mapas de Interação de Proteínas/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Catepsinas/antagonistas & inibidores , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/patologia , Camundongos , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Ratos , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
16.
Cells ; 9(9)2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32887380

RESUMO

Cathepsin S (CatS) is a secreted cysteine protease that cleaves certain extracellular matrix proteins, regulates antigen presentation in antigen-presenting cells (APC), and promotes M2-type macrophage and dendritic cell polarization. CatS is overexpressed in many solid cancers, and overall, it appears to promote an immune-suppressive and tumor-promoting microenvironment. While most data suggest that CatS inhibition or knockdown promotes anti-cancer immunity, cell-specific inhibition, especially in myeloid cells, appears to be important for therapeutic efficacy. This makes the design of CatS selective inhibitors and their targeting to tumor-associated M2-type macrophages (TAM) and DC an attractive therapeutic strategy compared to the use of non-selective immunosuppressive compounds or untargeted approaches. The selective inhibition of CatS can be achieved through optimized small molecule inhibitors that show good pharmacokinetic profiles and are orally bioavailable. The targeting of these inhibitors to TAM is now more feasible using nanocarriers that are functionalized for a directed delivery. This review discusses the role of CatS in the immunological tumor microenvironment and upcoming possibilities for a nanocarrier-mediated delivery of potent and selective CatS inhibitors to TAM and related APC to promote anti-tumor immunity.


Assuntos
Catepsinas/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Nanopartículas/administração & dosagem , Neoplasias/tratamento farmacológico , Inibidores de Proteases/farmacologia , Macrófagos Associados a Tumor/efeitos dos fármacos , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/genética , Antineoplásicos/síntese química , Antineoplásicos/farmacocinética , Azepinas/síntese química , Azepinas/farmacologia , Catepsinas/antagonistas & inibidores , Catepsinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/patologia , Dipeptídeos/síntese química , Dipeptídeos/farmacologia , Portadores de Fármacos/síntese química , Portadores de Fármacos/farmacocinética , Humanos , Imunoterapia/métodos , Leucina/análogos & derivados , Leucina/síntese química , Leucina/farmacologia , Terapia de Alvo Molecular/métodos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Inibidores de Proteases/síntese química , Sulfonas/síntese química , Sulfonas/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/patologia
17.
Bioorg Med Chem Lett ; 30(18): 127420, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32763808

RESUMO

A library of cathepsin S inhibitors of the dipeptide nitrile chemotype, bearing a bioisosteric sulfonamide moiety, was synthesized. Kinetic investigations were performed at four human cysteine proteases, i.e. cathepsins S, B, K and L. Compound 12 with a terminal 3-biphenyl sulfonamide substituent was the most potent (Ki = 4.02 nM; selectivity ratio cathepsin S/K = 5.8; S/L = 67) and 24 with a 4'-fluoro-4-biphenyl sulfonamide substituent the most selective cathepsin S inhibitor (Ki = 35.5 nM; selectivity ratio cathepsin S/K = 57; S/L = 31). In silico design and biochemical evaluation emphasized the impact of the sulfonamide linkage on selectivity and a possible switch of P2 and P3 substituents with respect to the occupation of the corresponding binding sites of cathepsin S.


Assuntos
Catepsinas/antagonistas & inibidores , Dipeptídeos/síntese química , Inibidores Enzimáticos/síntese química , Nitrilas/síntese química , Sulfonamidas/química , Sequência de Aminoácidos , Sítios de Ligação , Catepsina K/metabolismo , Catepsina L/metabolismo , Simulação por Computador , Cisteína Proteases/metabolismo , Humanos , Cinética , Ligação Proteica , Relação Estrutura-Atividade
18.
Int J Biol Macromol ; 164: 2745-2752, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32800952

RESUMO

Nano crystalline cellulose (NCC) modified with cationic surfactant CTAB (N-Cetyl-N, N, N-trimethyl ammonium bromide) proved as effective drug delivery excipient for sustained release of non-steroidal anti-inflammatory drugs (NSAIDs). In the present work, our purpose was to study the effect of another cationic surfactant TBAB (tetra-n-butyl ammonium bromide) used to modify NCC on binding and sustained release of hydrophobic NSAIDs. NCC modified TBAB was characterized by FTIR & TGA analysis. TBAB-NSAID interactions have been assessed using CHEM3D. The sustained release character was monitored by time dependent serum protein protecting and time dependent anti-cathepsins activities. The NCC-TBAB exhibited the binding efficiency for NSAIDs in the range of 42-25% depending upon the hydrophobicity of NSAID and released the drug up to 60 min. The research problem added to the existing knowledge of suitable surfactants required to change the hydrophilic nature of NCC to act as carrier for hydrophobic drugs. TBAB surfactant having shorter alkyl chain length exhibited lower binding efficiency and decreased sustain release of drugs as compared to CTAB having longer alkyl chain length.


Assuntos
Anti-Inflamatórios não Esteroides/química , Brometos/química , Celulose/química , Compostos de Amônio Quaternário/química , Tensoativos/química , Anti-Inflamatórios não Esteroides/farmacologia , Catepsinas/antagonistas & inibidores , Preparações de Ação Retardada , Sistemas de Liberação de Medicamentos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Nanopartículas , Fatores de Tempo
19.
Eur J Pharmacol ; 880: 173171, 2020 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-32437743

RESUMO

Cathepsin S (CatS) is a cysteine protease found in lysosomes of hematopoietic and microglial cells and in secreted form in the extracellular space. While CatS has been shown to contribute significantly to neuropathic pain, the precise mechanisms remain unclear. In this report, we describe JNJ-39641160, a novel non-covalent, potent, selective and orally-available CatS inhibitor that is peripherally restricted (non-CNS penetrant) and may represent an innovative class of immunosuppressive and analgesic compounds and tools useful toward investigating peripheral mechanisms of CatS in neuropathic pain. In C57BL/6 mice, JNJ-39641160 dose-dependently blocked the proteolysis of the invariant chain, and inhibited both T-cell activation and antibody production to a vaccine antigen. In the spared nerve injury (SNI) model of chronic neuropathic pain, in which T-cell activation has previously been demonstrated to be a prerequisite for the development of pain hypersensitivity, JNJ-39641160 fully reversed tactile allodynia in wild-type mice but was completely ineffective in the same model in CatS knockout mice (which exhibited a delayed onset in allodynia). By contrast, in the acute mild thermal injury (MTI) model, JNJ-39641160 only weakly attenuated allodynia at the highest dose tested. These findings support the hypothesis that blockade of peripheral CatS alone is sufficient to fully reverse allodynia following peripheral nerve injury and suggest that the mechanism of action likely involves interruption of T-cell activation and peripheral cytokine release. In addition, they provide important insights toward the development of selective CatS inhibitors for the treatment of neuropathic pain in humans.


Assuntos
Analgésicos/uso terapêutico , Catepsinas/antagonistas & inibidores , Hiperalgesia/tratamento farmacológico , Imunossupressores/uso terapêutico , Neuralgia/tratamento farmacológico , Traumatismos dos Nervos Periféricos/tratamento farmacológico , Inibidores de Proteases/uso terapêutico , Analgésicos/farmacocinética , Analgésicos/farmacologia , Animais , Encéfalo/metabolismo , Catepsinas/genética , Catepsinas/metabolismo , Linhagem Celular , Citocinas/imunologia , Temperatura Alta , Humanos , Hiperalgesia/imunologia , Imunoglobulina E/imunologia , Imunoglobulina G/imunologia , Imunossupressores/farmacocinética , Imunossupressores/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuralgia/imunologia , Traumatismos dos Nervos Periféricos/imunologia , Inibidores de Proteases/farmacocinética , Inibidores de Proteases/farmacologia , Nervo Isquiático/lesões , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Toxoide Tetânico/administração & dosagem , Tato
20.
Respir Res ; 21(1): 111, 2020 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-32398133

RESUMO

Dysregulated expression and activity of cathepsin S (CTSS), a lysosomal protease and a member of the cysteine cathepsin protease family, is linked to the pathogenesis of multiple diseases, including a number of conditions affecting the lungs. Extracellular CTSS has potent elastase activity and by processing cytokines and host defense proteins, it also plays a role in the regulation of inflammation. CTSS has also been linked to G-coupled protein receptor activation and possesses an important intracellular role in major histocompatibility complex class II antigen presentation. Modulated CTSS activity is also associated with pulmonary disease comorbidities, such as cancer, cardiovascular disease, and diabetes. CTSS is expressed in a wide variety of immune cells and is biologically active at neutral pH. Herein, we review the significance of CTSS signaling in pulmonary diseases and associated comorbidities. We also discuss CTSS as a plausible therapeutic target and describe recent and current clinical trials examining CTSS inhibition as a means for treatment.


Assuntos
Catepsinas/metabolismo , Mediadores da Inflamação/metabolismo , Pneumopatias/metabolismo , Pulmão/metabolismo , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/metabolismo , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/metabolismo , Catepsinas/antagonistas & inibidores , Ensaios Clínicos como Assunto/métodos , Comorbidade , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Pulmão/efeitos dos fármacos , Pneumopatias/tratamento farmacológico , Pneumopatias/epidemiologia , Neoplasias/tratamento farmacológico , Neoplasias/epidemiologia , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...