Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Pharmacol Ther ; 109(5): 1293-1303, 2021 05.
Article in English | MEDLINE | ID: mdl-33113155

ABSTRACT

Activated T cells drive a range of immune-mediated inflammatory diseases. LAG-3 is transiently expressed on recently activated CD4+ and CD8+ T cells. We describe the engineering and first-in-human clinical study (NCT02195349) of GSK2831781 (an afucosylated humanized IgG1 monoclonal antibody enhanced with high affinity for Fc receptors and LAG-3 and antibody-dependent cellular cytotoxicity capabilities), which depletes LAG-3 expressing cells. GSK2831781 was tested in a phase I/Ib, double-blind, placebo-controlled clinical study, which randomized 40 healthy participants (part A) and 27 patients with psoriasis (part B) to single doses of GSK2831781 (up to 0.15 and 5 mg/kg, respectively) or placebo. Adverse events were generally balanced across groups, with no safety or tolerability concern identified. LAG-3+ cell depletion in peripheral blood was observed at doses ≥ 0.15 mg/kg and was dose-dependent. In biopsies of psoriasis plaques, a reduction in mean group LAG-3+ and CD3+ T-cell counts was observed following treatment. Downregulation of proinflammatory genes (IL-17A, IL-17F, IFNγ, and S100A12) and upregulation of the epithelial barrier integrity gene, CDHR1, was observed with the 5 mg/kg dose of GSK2831781. Psoriasis disease activity improved up to day 43 at all GSK2831781 doses (0.5, 1.5, and 5 mg/kg) compared with placebo. Depletion of LAG-3-expressing activated T cells is a novel approach, and this first clinical study shows that GSK2831781 is pharmacologically active and provides encouraging early evidence of clinical effects in psoriasis, which warrants further investigation in T-cell-mediated inflammatory diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Psoriasis/drug therapy , T-Lymphocytes/immunology , Adult , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antigens, CD/blood , CD3 Complex/metabolism , Dose-Response Relationship, Immunologic , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Psoriasis/genetics , Psoriasis/pathology , Treatment Outcome , Lymphocyte Activation Gene 3 Protein
2.
Nat Cell Biol ; 14(2): 201-8, 2012 Jan 29.
Article in English | MEDLINE | ID: mdl-22286099

ABSTRACT

There are three prolyl hydroxylases (PHD1, 2 and 3) that regulate the hypoxia-inducible factors (HIFs), the master transcriptional regulators that respond to changes in intracellular O(2) tension. In high O(2) tension (normoxia) the PHDs hydroxylate two conserved proline residues on HIF-1α, which leads to binding of the von Hippel-Lindau (VHL) tumour suppressor, the recognition component of a ubiquitin-ligase complex, initiating HIF-1α ubiquitylation and degradation. However, it is not known whether PHDs and VHL act separately to exert their enzymatic activities on HIF-1α or as a multiprotein complex. Here we show that the tumour suppressor protein LIMD1 (LIM domain-containing protein) acts as a molecular scaffold, simultaneously binding the PHDs and VHL, thereby assembling a PHD-LIMD1-VHL protein complex and creating an enzymatic niche that enables efficient degradation of HIF-1α. Depletion of endogenous LIMD1 increases HIF-1α levels and transcriptional activity in both normoxia and hypoxia. Conversely, LIMD1 expression downregulates HIF-1 transcriptional activity in a manner depending on PHD and 26S proteasome activities. LIMD1 family member proteins Ajuba and WTIP also bind to VHL and PHDs 1 and 3, indicating that these LIM domain-containing proteins represent a previously unrecognized group of hypoxic regulators.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Procollagen-Proline Dioxygenase/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Cell Hypoxia , Cell Line, Tumor , HEK293 Cells , HeLa Cells , Humans , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases , Immunoblotting , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Models, Biological , Polyubiquitin/metabolism , Procollagen-Proline Dioxygenase/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , RNA Interference , Transfection , Two-Hybrid System Techniques , Ubiquitination , Von Hippel-Lindau Tumor Suppressor Protein/genetics
3.
Proc Natl Acad Sci U S A ; 107(28): 12499-504, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20616046

ABSTRACT

In recent years there have been major advances with respect to the identification of the protein components and mechanisms of microRNA (miRNA) mediated silencing. However, the complete and precise repertoire of components and mechanism(s) of action remain to be fully elucidated. Herein we reveal the identification of a family of three LIM domain-containing proteins, LIMD1, Ajuba and WTIP (Ajuba LIM proteins) as novel mammalian processing body (P-body) components, which highlight a novel mechanism of miRNA-mediated gene silencing. Furthermore, we reveal that LIMD1, Ajuba, and WTIP bind to Ago1/2, RCK, Dcp2, and eIF4E in vivo, that they are required for miRNA-mediated, but not siRNA-mediated gene silencing and that all three proteins bind to the mRNA 5' m(7)GTP cap-protein complex. Mechanistically, we propose the Ajuba LIM proteins interact with the m(7)GTP cap structure via a specific interaction with eIF4E that prevents 4EBP1 and eIF4G interaction. In addition, these LIM-domain proteins facilitate miRNA-mediated gene silencing by acting as an essential molecular link between the translationally inhibited eIF4E-m(7)GTP-5(')cap and Ago1/2 within the miRISC complex attached to the 3'-UTR of mRNA, creating an inhibitory closed-loop complex.


Subject(s)
Carrier Proteins/metabolism , Gene Silencing , MicroRNAs/metabolism , Animals , Carrier Proteins/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , Genes , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Mice , MicroRNAs/genetics , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
4.
Proc Natl Acad Sci U S A ; 105(50): 19932-7, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19060205

ABSTRACT

Loss of heterozygosity (LOH) and homozygous deletions at chromosome 3p21.3 are common in both small and nonsmall cell lung cancers, indicating the likely presence of tumor suppressor genes (TSGs). Although genetic and epigenetic changes within this region have been identified, the functional significance of these changes has not been explored. Concurrent protein expression and genetic analyses of human lung tumors coupled with functional studies have not been done. Here, we show that expression of the 3p21.3 gene, LIMD1, is frequently down-regulated in human lung tumors. Loss of LIMD1 expression occurs through a combination of gene deletion, LOH, and epigenetic silencing of transcription without evidence for coding region mutations. Experimentally, LIMD1 is a bona fide TSG. Limd1(-/-) mice are predisposed to chemical-induced lung adenocarcinoma and genetic inactivation of Limd1 in mice heterozygous for oncogenic K-Ras(G12D) markedly increased tumor initiation, promotion, and mortality. Thus, we conclude that LIMD1 is a validated chromosome 3p21.3 tumor-suppressor gene involved in human lung cancer development. LIMD1 is a LIM domain containing adapter protein that localizes to E-cadherin cell-cell adhesive junctions, yet also translocates to the nucleus where it has been shown to function as an RB corepressor. As such, LIMD1 has the potential to communicate cell extrinsic or environmental cues with nuclear responses.


Subject(s)
Adenocarcinoma/genetics , Chromosomes, Human, Pair 3/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Adenocarcinoma/chemically induced , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , DNA Methylation , Gene Deletion , Gene Silencing , Genes, ras , Genetic Predisposition to Disease , Humans , Intracellular Signaling Peptides and Proteins/physiology , LIM Domain Proteins , Loss of Heterozygosity , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mice , Mice, Mutant Strains , Promoter Regions, Genetic , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology
5.
Int J Cancer ; 123(10): 2247-53, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18712738

ABSTRACT

The tumour suppressor gene (TSG) LIM domain containing protein 1 (LIMD1) has been associated with transformation of epithelial cells of the lung and its expression is downregulated in all lung tumour samples tested compared to normal lung matched controls. In the first study of its kind we used an anti-LIMD1 specific monoclonal antibody to investigate expression/localisation of the LIMD1 protein in a well-characterised tissue microarray of breast cancers and normal adjacent epithelia. Comparison of tumour with adjacent normal and distant normal tissue demonstrated that LIMD1 expression is moderate to high compared to tumour. There was also a significant correlation with histological grade (p = 0.0001), tumour size (p = 0.013) and tumour type (p = 0.004) indicating an association with aggressive disease. Cytoplasmic LIMD1 expression was seen in 99.3% of cases, with 43.1% showing both nuclear and cytoplasmic localisation. Absence/loss of nuclear staining showed a strong correlation with patient survival and was indicative of poor prognosis (p = 0.033). There was no association with lymph node status and other clinicopathological parameters. Nuclear staining was more pronounced in better prognosis tumours and normal tissue. This study demonstrates that LIMD1 represents a novel prognostic marker for breast cancer. Combined with the fact that LIMD1 expression is downregulated in lung cancers this clearly indicates that LIMD1 may represent a critical TSG, the function of which is deregulated via overall loss of expression and/or relocalisation within the cell during tumour development. The possible functions of LIMD1 localisation within the nucleus and cytoplasm and its relationship to tumour prognosis are discussed.


Subject(s)
Breast Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Subcellular Fractions/metabolism , Survival , Adult , Aged , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Immunohistochemistry , LIM Domain Proteins , Middle Aged , Neoplasm Invasiveness
SELECTION OF CITATIONS
SEARCH DETAIL
...