Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Eur Respir Rev ; 32(168)2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37137509

ABSTRACT

Clinical management of cystic fibrosis (CF) has been greatly improved by the development of small molecule modulators of the CF transmembrane conductance regulator (CFTR). These drugs help to address some of the basic genetic defects of CFTR; however, no suitable CFTR modulators exist for 10% of people with CF (PWCF). An alternative, mutation-agnostic therapeutic approach is therefore still required. In CF airways, elevated levels of the proprotein convertase furin contribute to the dysregulation of key processes that drive disease pathogenesis. Furin plays a critical role in the proteolytic activation of the epithelial sodium channel; hyperactivity of which causes airways dehydration and loss of effective mucociliary clearance. Furin is also responsible for the processing of transforming growth factor-ß, which is increased in bronchoalveolar lavage fluid from PWCF and is associated with neutrophilic inflammation and reduced pulmonary function. Pathogenic substrates of furin include Pseudomonas exotoxin A, a major toxic product associated with Pseudomonas aeruginosa infection and the spike glycoprotein of severe acute respiratory syndrome coronavirus 2, the causative pathogen for coronavirus disease 2019. In this review we discuss the importance of furin substrates in the progression of CF airways disease and highlight selective furin inhibition as a therapeutic strategy to provide clinical benefit to all PWCF.


Subject(s)
COVID-19 , Cystic Fibrosis , Humans , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Furin/pharmacology , Furin/therapeutic use , Mucociliary Clearance
2.
Br J Cancer ; 128(7): 1189-1195, 2023 03.
Article in English | MEDLINE | ID: mdl-36522477

ABSTRACT

Immunotherapy is becoming an advanced clinical management for various cancers. Rebuilding of aberrant immune surveillance on cancers has achieved notable progress in the past years by either in vivo or ex vivo engineering of efficient immune cells. Immune cells can be programmed with several strategies that improves their therapeutic influence and specificity. It has become noticeable that effective immunotherapy must consider the complete complexity of the immune cell function. However, today, almost all immune cells can be transiently or stably reprogrammed against various cancer cells. As a consequence, investigations have interrogated strategies to improve the efficacy of cancer immunotherapies by enhancing T-cell infiltration into tumour tissues. Here, we review the emerging role of furin-like enzymes work related to T-cell reprogramming, their tumour infiltration and cytotoxic function.


Subject(s)
Furin , Neoplasms , Humans , Furin/therapeutic use , Immunotherapy , Neoplasms/drug therapy , T-Lymphocytes/pathology , Tumor Microenvironment
3.
Br J Cancer ; 122(6): 885-894, 2020 03.
Article in English | MEDLINE | ID: mdl-31988347

ABSTRACT

BACKGROUND: Breast cancer is the second most common cancer in the world. Despite advances in therapies, the mechanisms of resistance remain the underlying cause of morbidity and mortality. Lipoic acid (LA) is an antioxidant and essential cofactor in oxidative metabolism. Its potential therapeutic effects have been well documented, but its mechanisms of action (MOA) are not fully understood. METHODS: The aim of this study is to validate the inhibitory LA effect on the proliferation of various breast cancer cell lines and to investigate the MOA that may be involved in this process. We tested LA effects by ex vivo studies on fresh human mammary tumour samples. RESULTS: We demonstrate that LA inhibits the proliferation and Akt and ERK signalling pathways of several breast cancer cells. While searching for upstream dysregulations, we discovered the loss of expression of IGF-1R upon exposure to LA. This decrease is due to the downregulation of the convertase, furin, which is implicated in the maturation of IGF-1R. Moreover, ex vivo studies on human tumour samples showed that LA significantly decreases the expression of the proliferation marker Ki67. CONCLUSION: LA exerts its anti-proliferative effect by inhibiting the maturation of IGF-1R via the downregulation of furin.


Subject(s)
Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Furin/therapeutic use , Receptor, IGF Type 1/antagonists & inhibitors , Thioctic Acid/therapeutic use , Breast Neoplasms/pathology , Down-Regulation , Female , Furin/pharmacology , Humans , Thioctic Acid/pharmacology , Transfection
4.
Hum Mol Genet ; 26(7): 1353-1364, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28334940

ABSTRACT

Gelsolin amyloidosis is a dominantly inherited, incurable type of amyloidosis. A single point mutation in the gelsolin gene (G654A is most common) results in the loss of a Ca2+ binding site in the second gelsolin domain. Consequently, this domain partly unfolds and exposes an otherwise buried furin cleavage site at the surface. During secretion of mutant plasma gelsolin consecutive cleavage by furin and MT1-MMP results in the production of 8 and 5 kDa amyloidogenic peptides. Nanobodies that are able to (partly) inhibit furin or MT1-MMP proteolysis have previously been reported. In this study, the nanobodies have been combined into a single bispecific format able to simultaneously shield mutant plasma gelsolin from intracellular furin and extracellular MT1-MMP activity. We report the successful in vivo expression of this bispecific nanobody following adeno-associated virus serotype 9 gene therapy in gelsolin amyloidosis mice. Using SPECT/CT and immunohistochemistry, a reduction in gelsolin amyloid burden was detected which translated into improved muscle contractile properties. We conclude that a nanobody-based gene therapy using adeno-associated viruses shows great potential as a novel strategy in gelsolin amyloidosis and potentially other amyloid diseases.


Subject(s)
Amyloidosis/genetics , Amyloidosis/therapy , Gelsolin/genetics , Genetic Therapy , Amyloidosis/pathology , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/therapeutic use , Dependovirus/genetics , Dependovirus/immunology , Disease Models, Animal , Furin/immunology , Furin/therapeutic use , Gelsolin/immunology , Humans , Matrix Metalloproteinase 14/immunology , Matrix Metalloproteinase 14/therapeutic use , Mice , Point Mutation/genetics , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology
5.
Expert Opin Ther Pat ; 25(4): 379-96, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25563687

ABSTRACT

INTRODUCTION: Since the discovery of furin, numerous reports have studied its role in health and diseases, including cancer, inflammatory and infectious diseases. This interest has led to the development of both large protein- and peptide-based inhibitors aiming to control furin activity to treat these disorders. The most recent advances include the development of potent peptidomimetic furin inhibitors, considerably expanding the field of therapeutic applications. AREA COVERED: In this review, the use of furin or its inhibitors for therapeutic conditions is described through the patent literature since 1994. Only compounds with biological efficacy or augmented properties demonstrated within the patent literature or the associated publications concerning their claimed uses are discussed. EXPERT OPINION: Considering the diseases that may benefit from furin inhibition, several patents detail the use of the restricted number of furin inhibitors. However, there have been recent reports of new scaffolds, and even the use of furin itself, as a therapeutic agent. Despite considerable evidence of in vivo efficacy, limited confirmation from clinical trials supports or refutes the further use of these compounds in a therapeutic context. The most advanced application is the use of furin knockdown in the generation of an autologous cancer vaccine, which has initiated clinical trials.


Subject(s)
Drug Design , Furin/metabolism , Peptidomimetics/pharmacology , Animals , Cancer Vaccines/pharmacology , Clinical Trials as Topic , Furin/antagonists & inhibitors , Furin/therapeutic use , Gene Knockdown Techniques , Humans , Patents as Topic
6.
Arthritis Rheum ; 64(9): 2878-86, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22605541

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) is an autoimmune joint disease associated with chronic inflammation of the synovium that causes profound damage of joints. Inflammation results in part from the influx of immune cells secreting inflammatory cytokines and the reduction in the number of Treg cells. We undertook this study to assess the effect of furin, a proteinase implicated in the proteolytic activity of various precursor proteins and involved in the regulation of both proteinase maturation and immune cells, in an experimental model of RA. METHODS: The effect of furin and its inhibitor α1-PDX was tested in mice with collagen-induced arthritis (CIA). Joints were processed for histology and protein expression. Levels of cytokines were measured in joint tissue, and Treg cell numbers were measured in spleens. RESULTS: Furin expression and activity were high in the synovial pannus in RA patients and mice with CIA. Systemic administration of furin prevented increases in the arthritis score, joint destruction, and bone loss, in contrast to systemic administration of the furin inhibitor α1-PDX, which enhanced these parameters. By preventing the development of synovial pannus, furin reduced the expression of metalloproteinases in the joints. In contrast, α1-PDX enhanced synovial proliferation and the expression and activity of matrix metalloproteinases. Furthermore, furin reversed the local Th1/Th2 balance and restored the number of Treg cells in the spleen, indicating mediation by immune cells. CONCLUSION: These findings show the protective role of exogenous furin against RA, mediated by an immune response. The data suggest the potential therapeutic use of furin or its derivatives in autoimmune diseases including RA.


Subject(s)
Arthritis, Experimental/immunology , Furin/pharmacology , Joints/drug effects , Rheumatic Fever/immunology , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Cytokines/metabolism , Furin/therapeutic use , Humans , Joints/metabolism , Joints/pathology , Male , Mice , Mice, Inbred DBA , Rheumatic Fever/drug therapy , Rheumatic Fever/pathology , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Synovial Membrane/pathology , alpha 1-Antitrypsin/pharmacology
7.
Mol Ther ; 9(5): 674-81, 2004 May.
Article in English | MEDLINE | ID: mdl-15120328

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to exert selectively cytotoxic activity against many tumor cells but not normal cells. On the other hand, the ligand for the receptor tyrosine kinase Fms-like tyrosine kinase 3 (Flt3L) is a growth factor for hematopoietic progenitors and is a potent stimulating factor for dendritic and NK cells. Previously, we have demonstrated that it is possible to inhibit the outgrowth of primary tumors by the administration of an hFlex (the extracellular domain of the Flt3L) and TRAIL (amino acid residues 95-281) secreted fusion protein. Here, we report that by the insertion of a linker sequence encoding the cleavage site for the Golgi-expressed endoprotease furin between the DNA sequences encoding hFlex and TRAIL, the tumoricidal activity of the cleaved TRAIL protein generated was greatly enhanced in comparison to the hFlex/TRAIL fusion protein. Furthermore, we demonstrate that intratumoral injection of the hFlex/furin/TRAIL DNA, in conjunction with cationic liposomes, significantly suppressed the outgrowth of the human CNE-2 nasopharyngeal tumor xenografts in SCID mice. In situ histological examinations confirmed the expression of TRAIL in the treated tumor nodules and the induction of apoptosis was also evidenced by the presence of numerous pyknotic nuclei.


Subject(s)
Furin/genetics , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Neoplasms/therapy , Tumor Necrosis Factor-alpha/genetics , Animals , Annexin A5/analysis , Antibodies/immunology , Apoptosis , Apoptosis Regulatory Proteins , Carcinoma/pathology , Carcinoma/therapy , Cell Line, Tumor , DNA, Recombinant/genetics , DNA, Recombinant/therapeutic use , Dendritic Cells/immunology , Furin/therapeutic use , Humans , Immunochemistry , Liposomes/therapeutic use , Membrane Proteins/therapeutic use , Mice , Mice, SCID , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/therapy , Neoplasm Transplantation , Plasmids/genetics , Plasmids/therapeutic use , Propidium/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , TNF-Related Apoptosis-Inducing Ligand , Transfection , Transplantation, Heterotopic , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...