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.
Aliment Pharmacol Ther ; 58(3): 283-296, 2023 08.
Article in English | MEDLINE | ID: mdl-37323059

ABSTRACT

BACKGROUND: Selective depletion of T cells expressing LAG-3, an immune checkpoint receptor that is upregulated on activated T cells, has been investigated in pre-clinical models as a potential therapeutic approach in inflammatory and autoimmune diseases where activated T cells are implicated. AIMS: GSK2831781, a depleting monoclonal antibody that specifically binds LAG-3 proteins, may deplete activated LAG-3+ cells in ulcerative colitis (UC). METHODS: Patients with moderate to severe UC were randomised to GSK2831781 or placebo. Safety, tolerability, efficacy, pharmacokinetics and pharmacodynamics of GSK2831781 were evaluated. RESULTS: One hundred four participants across all dose levels were randomised prior to an interim analysis indicating efficacy futility criteria had been met. Efficacy results focus on the double-blind induction phase of the study (GSK2831781 450 mg intravenously [IV], N = 48; placebo, N = 27). Median change from baseline (95% credible interval [CrI]) in complete Mayo score was similar between groups (GSK2831781 450 mg IV: -1.4 [-2.2, -0.7]; placebo: -1.4 [-2.4, -0.5]). Response rates for endoscopic improvement favoured placebo. Clinical remission rates were similar between groups. In the 450-mg IV group, 14 (29%) participants had an adverse event of UC versus 1 (4%) with placebo. LAG-3+ cells were depleted to 51% of baseline in blood; however, there was no reduction in LAG-3+ cells in the colonic mucosa. Transcriptomic analysis of colon biopsies showed no difference between groups. CONCLUSION: Despite evidence of target cell depletion in blood, GSK2831781 failed to reduce inflammation in the colonic mucosa suggesting no pharmacological effect. The study was terminated early (NCT03893565).


Subject(s)
Colitis, Ulcerative , Humans , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Antibodies, Monoclonal/adverse effects , Double-Blind Method , T-Lymphocytes , Remission Induction , Treatment Outcome
2.
Front Immunol ; 13: 795463, 2022.
Article in English | MEDLINE | ID: mdl-35197971

ABSTRACT

Protection from liver-stage malaria requires high numbers of CD8+ T cells to find and kill Plasmodium-infected cells. A new malaria vaccine strategy, prime-target vaccination, involves sequential viral-vectored vaccination by intramuscular and intravenous routes to target cellular immunity to the liver. Liver tissue-resident memory (TRM) CD8+ T cells have been shown to be necessary and sufficient for protection against rodent malaria by this vaccine regimen. Ultimately, to most faithfully assess immunotherapeutic responses by these local, specialised, hepatic T cells, periodic liver sampling is necessary, however this is not feasible at large scales in human trials. Here, as part of a phase I/II P. falciparum challenge study of prime-target vaccination, we performed deep immune phenotyping, single-cell RNA-sequencing and kinetics of hepatic fine needle aspirates and peripheral blood samples to study liver CD8+ TRM cells and circulating counterparts. We found that while these peripheral 'TRM-like' cells differed to TRM cells in terms of previously described characteristics, they are similar phenotypically and indistinguishable in terms of key T cell residency transcriptional signatures. By exploring the heterogeneity among liver CD8+ TRM cells at single cell resolution we found two main subpopulations that each share expression profiles with blood T cells. Lastly, our work points towards the potential for using TRM-like cells as a correlate of protection by liver-stage malaria vaccines and, in particular, those adopting a prime-target approach. A simple and reproducible correlate of protection would be particularly valuable in trials of liver-stage malaria vaccines as they progress to phase III, large-scale testing in African infants. We provide a blueprint for understanding and monitoring liver TRM cells induced by a prime-target malaria vaccine approach.


Subject(s)
Malaria Vaccines/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors , Hepatocytes/immunology , Humans , Immunity, Cellular , Immunologic Memory/immunology , Liver/immunology , Malaria/immunology , Plasmodium/immunology , Sporozoites/immunology , Transcriptome , Vaccination
3.
Front Oncol ; 11: 779739, 2021.
Article in English | MEDLINE | ID: mdl-34900733

ABSTRACT

Hypoxia is a common phenomenon in solid tumours strongly linked to the hallmarks of cancer. Hypoxia promotes local immunosuppression and downregulates type I interferon (IFN) expression and signalling, which contribute to the success of many cancer therapies. Double-stranded RNA (dsRNA), transiently generated during mitochondrial transcription, endogenously activates the type I IFN pathway. We report the effects of hypoxia on the generation of mitochondrial dsRNA (mtdsRNA) in breast cancer. We found a significant decrease in dsRNA production in different cell lines under hypoxia. This effect was HIF1α/2α-independent. mtdsRNA was responsible for induction of type I IFN and significantly decreased after hypoxia. Mitochondrially encoded gene expression was downregulated and mtdsRNA bound by the dsRNA-specific J2 antibody was decreased during hypoxia. These findings reveal a new mechanism of hypoxia-induced immunosuppression that could be targeted by hypoxia-activated therapies.

4.
BMC Cancer ; 21(1): 896, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34353313

ABSTRACT

BACKGROUND: In clear cell renal cell carcinoma, 80% of cases have biallelic inactivation of the VHL gene, leading to constitutive activation of both HIF1α and HIF2α. As HIF2α is the driver of the disease promoting tumour growth and metastasis, drugs targeting HIF2α have been developed. However, resistance is common, therefore new therapies are needed. METHODS: We assessed the effect of the HIF2α antagonist PT2385 in several steps of tumour development and performed RNAseq to identify genes differentially expressed upon treatment. A drug screening was used to identify drugs with antiproliferative effects on VHL-mutated HIF2α-expressing cells and could increase effectiveness of PT2385. RESULTS: PT2385 did not reduce cell proliferation or clonogenicity but, in contrast to the genetic silencing of HIF2α, it reduced in vitro cell invasion. Many HIF-inducible genes were down-regulated upon PT2385 treatment, whereas some genes involved in cell migration or extracellular matrix were up-regulated. HIF2α was associated with resistance to statins, addition to PT2385 did not increase the sensitivity. CONCLUSIONS: this study shows key differences between inhibiting a target versus knockdown, which are potentially targetable.


Subject(s)
Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Silencing , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Repositioning , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Humans , Indans/pharmacology , Indans/therapeutic use , Kidney Neoplasms/drug therapy , Sulfones/pharmacology , Sulfones/therapeutic use , Transcriptional Activation , Transcriptome , Treatment Outcome
5.
Cell Rep ; 24(12): 3194-3206.e4, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30232002

ABSTRACT

A balanced diet of macronutrients is critical for animal health. A lack of specific elements can have profound effects on behavior, reproduction, and lifespan. Here, we used Drosophila to understand how the brain responds to carbohydrate deprivation. We found that serine protease homologs (SPHs) are enriched among genes that are transcriptionally regulated in flies deprived of carbohydrates. Stimulation of neurons expressing one of these SPHs, Scarface (Scaf), or overexpression of scaf positively regulates feeding on nutritious sugars, whereas inhibition of these neurons or knockdown of scaf reduces feeding. This modulation of food intake occurs only in sated flies while hunger-induced feeding is unaffected. Furthermore, scaf expression correlates with the presence of sugar in the food. As Scaf and Scaf neurons promote feeding independent of the hunger state, and the levels of scaf are positively regulated by the presence of sugar, we conclude that scaf mediates the hedonic control of feeding.


Subject(s)
Brain/metabolism , Carbohydrate Metabolism , Drosophila Proteins/metabolism , Feeding Behavior , Serine Proteases/metabolism , Animals , Brain/drug effects , Dietary Carbohydrates/pharmacology , Drosophila Proteins/genetics , Drosophila melanogaster , Neurons/drug effects , Neurons/metabolism , Serine Proteases/genetics
6.
Sci Rep ; 6: 27885, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27296678

ABSTRACT

In the fruitfly Drosophila melanogaster, the differential development of wing and haltere is dependent on the function of the Hox protein Ultrabithorax (Ubx). Here we compare Ubx-mediated regulation of wing patterning genes between the honeybee, Apis mellifera, the silkmoth, Bombyx mori and Drosophila. Orthologues of Ubx are expressed in the third thoracic segment of Apis and Bombyx, although they make functional hindwings. When over-expressed in transgenic Drosophila, Ubx derived from Apis or Bombyx could suppress wing development, suggesting evolutionary changes at the level of co-factors and/or targets of Ubx. To gain further insights into such events, we identified direct targets of Ubx from Apis and Bombyx by ChIP-seq and compared them with those of Drosophila. While majority of the putative targets of Ubx are species-specific, a considerable number of wing-patterning genes are retained, over the past 300 millions years, as targets in all the three species. Interestingly, many of these are differentially expressed only between wing and haltere in Drosophila but not between forewing and hindwing in Apis or Bombyx. Detailed bioinformatics and experimental validation of enhancer sequences suggest that, perhaps along with other factors, changes in the cis-regulatory sequences of earlier targets contribute to diversity in Ubx function.


Subject(s)
Bees , Bombyx , Drosophila Proteins/metabolism , Drosophila melanogaster , Enhancer Elements, Genetic/genetics , Homeodomain Proteins/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/metabolism , Wings, Animal/physiology , Animals , Animals, Genetically Modified , Biological Evolution , Comparative Genomic Hybridization , Computational Biology , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Organogenesis , Species Specificity , Transcription Factors/genetics
7.
Int J Dev Biol ; 59(4-6): 187-94, 2015.
Article in English | MEDLINE | ID: mdl-26505251

ABSTRACT

Notch signalling is essential for animal development. It integrates multiple pathways controlling cell fate and specification. Here we report the genetic characterization of Gliolectin, presumably a lectin, a cytoplasmic protein, significantly enriched in Golgi bodies. Its expression overlaps with regions where Notch is activated. Loss of gliolectin function results in ectopic veins, while gain of its function causes loss of wing veins. It positively regulates Enhancer of split mß, a target of Notch signalling. These observations suggest that it is a positive regulator of Notch signalling during wing development in Drosophila.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Lectins/genetics , Nerve Tissue Proteins/genetics , Receptors, Notch/genetics , Signal Transduction/genetics , Wings, Animal/metabolism , Animals , Animals, Genetically Modified , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Golgi Apparatus/metabolism , Imaginal Discs/embryology , Imaginal Discs/growth & development , Imaginal Discs/metabolism , In Situ Hybridization , Lectins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Nerve Tissue Proteins/metabolism , RNA Interference , Receptors, Notch/metabolism , Wings, Animal/embryology , Wings, Animal/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...