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










Publication year range
1.
Mol Metab ; 84: 101955, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38704026

ABSTRACT

OBJECTIVE: The contribution of the mitochondrial electron transfer system to insulin secretion involves more than just energy provision. We identified a small RNA fragment (mt-tRF-LeuTAA) derived from the cleavage of a mitochondrially-encoded tRNA that is conserved between mice and humans. The role of mitochondrially-encoded tRNA-derived fragments remains unknown. This study aimed to characterize the impact of mt-tRF-LeuTAA, on mitochondrial metabolism and pancreatic islet functions. METHODS: We used antisense oligonucleotides to reduce mt-tRF-LeuTAA levels in primary rat and human islet cells, as well as in insulin-secreting cell lines. We performed a joint transcriptome and proteome analysis upon mt-tRF-LeuTAA inhibition. Additionally, we employed pull-down assays followed by mass spectrometry to identify direct interactors of the fragment. Finally, we characterized the impact of mt-tRF-LeuTAA silencing on the coupling between mitochondrial metabolism and insulin secretion using high-resolution respirometry and insulin secretion assays. RESULTS: Our study unveils a modulation of mt-tRF-LeuTAA levels in pancreatic islets in different Type 2 diabetes models and in response to changes in nutritional status. The level of the fragment is finely tuned by the mechanistic target of rapamycin complex 1. Located within mitochondria, mt-tRF-LeuTAA interacts with core subunits and assembly factors of respiratory complexes of the electron transfer system. Silencing of mt-tRF-LeuTAA in islet cells limits the inner mitochondrial membrane potential and impairs mitochondrial oxidative phosphorylation, predominantly by affecting the Succinate (via Complex II)-linked electron transfer pathway. Lowering mt-tRF-LeuTAA impairs insulin secretion of rat and human pancreatic ß-cells. CONCLUSIONS: Our findings indicate that mt-tRF-LeuTAA interacts with electron transfer system complexes and is a pivotal regulator of mitochondrial oxidative phosphorylation and its coupling to insulin secretion.


Subject(s)
Insulin Secretion , Insulin-Secreting Cells , Mitochondria , Animals , Rats , Humans , Mitochondria/metabolism , Insulin-Secreting Cells/metabolism , RNA, Transfer/metabolism , RNA, Transfer/genetics , Male , Insulin/metabolism , Islets of Langerhans/metabolism , Diabetes Mellitus, Type 2/metabolism , RNA, Mitochondrial/metabolism , RNA, Mitochondrial/genetics , Mice , Rats, Wistar , Electron Transport
2.
Diabetologia ; 66(7): 1306-1321, 2023 07.
Article in English | MEDLINE | ID: mdl-36995380

ABSTRACT

AIMS/HYPOTHESIS: Wolfram syndrome is a rare autosomal recessive disorder caused by pathogenic variants in the WFS1 gene. It is characterised by insulin-dependent diabetes mellitus, optic nerve atrophy, diabetes insipidus, hearing loss and neurodegeneration. Considering the unmet treatment need for this orphan disease, this study aimed to evaluate the therapeutic potential of glucagon-like peptide 1 receptor (GLP-1R) agonists under wolframin (WFS1) deficiency with a particular focus on human beta cells and neurons. METHODS: The effect of the GLP-1R agonists dulaglutide and exenatide was examined in Wfs1 knockout mice and in an array of human preclinical models of Wolfram syndrome, including WFS1-deficient human beta cells, human induced pluripotent stem cell (iPSC)-derived beta-like cells and neurons from control individuals and individuals affected by Wolfram syndrome, and humanised mice. RESULTS: Our study shows that the long-lasting GLP-1R agonist dulaglutide reverses impaired glucose tolerance in WFS1-deficient mice, and that exenatide and dulaglutide improve beta cell function and prevent apoptosis in different human WFS1-deficient models including iPSC-derived beta cells from people with Wolfram syndrome. Exenatide improved mitochondrial function, reduced oxidative stress and prevented apoptosis in Wolfram syndrome iPSC-derived neural precursors and cerebellar neurons. CONCLUSIONS/INTERPRETATION: Our study provides novel evidence for the beneficial effect of GLP-1R agonists on WFS1-deficient human pancreatic beta cells and neurons, suggesting that these drugs may be considered as a treatment for individuals with Wolfram syndrome.


Subject(s)
Induced Pluripotent Stem Cells , Insulin-Secreting Cells , Optic Atrophy , Wolfram Syndrome , Humans , Animals , Mice , Wolfram Syndrome/drug therapy , Wolfram Syndrome/genetics , Exenatide/therapeutic use , Optic Atrophy/pathology , Insulin-Secreting Cells/pathology , Mice, Knockout
3.
Cell Rep ; 40(2): 111069, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35830789

ABSTRACT

tRNA-derived fragments (tRFs) are an emerging class of small non-coding RNAs with distinct cellular functions. Here, we studied the contribution of tRFs to the regulation of postnatal ß cell maturation, a critical process that may lead to diabetes susceptibility in adulthood. We identified three tRFs abundant in neonatal rat islets originating from 5' halves (tiRNA-5s) of histidine and glutamate tRNAs. Their inhibition in these islets reduced ß cell proliferation and insulin secretion. Mitochondrial respiration was also perturbed, fitting with the mitochondrial enrichment of nuclear-encoded tiRNA-5HisGTG and tiRNA-5GluCTC. Notably, tiRNA-5 inhibition reduced Mpc1, a mitochondrial pyruvate carrier whose knock down largely phenocopied tiRNA-5 inhibition. tiRNA-5HisGTG interactome revealed binding to Musashi-1, which was essential for the mitochondrial enrichment of tiRNA-5HisGTG. Finally, tiRNA-5s were dysregulated in the islets of diabetic and diabetes-prone animals. Altogether, tiRNA-5s represent a class of regulators of ß cell maturation, and their deregulation in neonatal islets may lead to diabetes susceptibility in adulthood.


Subject(s)
Insulin-Secreting Cells , RNA, Transfer , Animals , Cell Proliferation , Insulin Secretion , Insulin-Secreting Cells/metabolism , RNA/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Rats
4.
Islets ; 13(3-4): 51-65, 2021 07 04.
Article in English | MEDLINE | ID: mdl-34241569

ABSTRACT

Exposure of human pancreatic beta cells to pro-inflammatory cytokines or metabolic stressors is used to model events related to type 1 and type 2 diabetes, respectively. Quantitative real-time PCR is commonly used to quantify changes in gene expression. The selection of the most adequate reference gene(s) for gene expression normalization is an important pre-requisite to obtain accurate and reliable results. There are no universally applicable reference genes, and the human beta cell expression of commonly used reference genes can be altered by different stressors. Here we aimed to identify the most stably expressed genes in human beta cells to normalize quantitative real-time PCR gene expression.We used comprehensive RNA-sequencing data from the human pancreatic beta cell line EndoC-ßH1, human islets exposed to cytokines or the free fatty acid palmitate in order to identify the most stably expressed genes. Genes were filtered based on their level of significance (adjusted P-value >0.05), fold-change (|fold-change| <1.5) and a coefficient of variation <10%. Candidate reference genes were validated by quantitative real-time PCR in independent samples.We identified a total of 264 genes stably expressed in EndoC-ßH1 cells and human islets following cytokines - or palmitate-induced stress, displaying a low coefficient of variation. Validation by quantitative real-time PCR of the top five genes ARF1, CWC15, RAB7A, SIAH1 and VAPA corroborated their expression stability under most of the tested conditions. Further validation in independent samples indicated that the geometric mean of ACTB and VAPA expression can be used as a reliable normalizing factor in human beta cells.


Subject(s)
Genomics/methods , Insulin-Secreting Cells , Humans , Insulin-Secreting Cells/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
5.
Int J Mol Sci ; 22(4)2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33578952

ABSTRACT

Macrophages are highly heterogeneous and plastic immune cells with peculiar characteristics dependent on their origin and microenvironment. Following pathogen infection or damage, circulating monocytes can be recruited in different tissues where they differentiate into macrophages. Stimuli present in the surrounding milieu induce the polarisation of macrophages towards a pro-inflammatory or anti-inflammatory profile, mediating inflammatory or homeostatic responses, respectively. However, macrophages can also derive from embryonic hematopoietic precursors and reside in specific tissues, actively participating in the development and the homeostasis in physiological conditions. Pancreatic islet resident macrophages are present from the prenatal stages onwards and show specific surface markers and functions. They localise in close proximity to ß-cells, being exquisite sensors of their secretory ability and viability. Over the years, the crucial role of macrophages in ß-cell differentiation and homeostasis has been highlighted. In addition, macrophages are emerging as central players in the initiation of autoimmune insulitis in type 1 diabetes and in the low-grade chronic inflammation characteristic of obesity and type 2 diabetes pathogenesis. The present work reviews the current knowledge in the field, with a particular focus on the mechanisms of communication between ß-cells and macrophages that have been described so far.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/immunology , Insulin-Secreting Cells/immunology , Macrophages/immunology , Animals , Cell Proliferation , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/pathology , Homeostasis , Humans , Inflammation/immunology , Inflammation/pathology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/pathology , Macrophages/cytology , Macrophages/pathology
6.
Eur J Endocrinol ; 184(3): 455-468, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33486469

ABSTRACT

OBJECTIVE: DNAJC3, also known as P58IPK, is an Hsp40 family member that interacts with and inhibits PKR-like ER-localized eIF2α kinase (PERK). Dnajc3 deficiency in mice causes pancreatic ß-cell loss and diabetes. Loss-of-function mutations in DNAJC3 cause early-onset diabetes and multisystemic neurodegeneration. The aim of our study was to investigate the genetic cause of early-onset syndromic diabetes in two unrelated patients, and elucidate the mechanisms of ß-cell failure in this syndrome. METHODS: Whole exome sequencing was performed and identified variants were confirmed by Sanger sequencing. DNAJC3 was silenced by RNAi in INS-1E cells, primary rat ß-cells, human islets, and induced pluripotent stem cell-derived ß-cells. ß-cell function and apoptosis were assessed, and potential mediators of apoptosis examined. RESULTS: The two patients presented with juvenile-onset diabetes, short stature, hypothyroidism, neurodegeneration, facial dysmorphism, hypoacusis, microcephaly and skeletal bone deformities. They were heterozygous compound and homozygous for novel loss-of-function mutations in DNAJC3. DNAJC3 silencing did not impair insulin content or secretion. Instead, the knockdown induced rat and human ß-cell apoptosis and further sensitized cells to endoplasmic reticulum stress, triggering mitochondrial apoptosis via the pro-apoptototic Bcl-2 proteins BIM and PUMA. CONCLUSIONS: This report confirms previously described features and expands the clinical spectrum of syndromic DNAJC3 diabetes, one of the five monogenic forms of diabetes pertaining to the PERK pathway of the endoplasmic reticulum stress response. DNAJC3 deficiency may lead to ß-cell loss through BIM- and PUMA-dependent activation of the mitochondrial pathway of apoptosis.


Subject(s)
Apoptosis/genetics , Diabetes Mellitus, Type 1/genetics , HSP40 Heat-Shock Proteins/genetics , Insulin-Secreting Cells/physiology , Mitochondria/metabolism , Adolescent , Adult , Age Factors , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Female , Humans , Insulin-Secreting Cells/metabolism , Loss of Function Mutation , Male , Mice , Mitochondria/pathology , Pedigree , Rats , Syndrome
7.
J Clin Invest ; 130(12): 6338-6353, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33164986

ABSTRACT

Neonatal diabetes is caused by single gene mutations reducing pancreatic ß cell number or impairing ß cell function. Understanding the genetic basis of rare diabetes subtypes highlights fundamental biological processes in ß cells. We identified 6 patients from 5 families with homozygous mutations in the YIPF5 gene, which is involved in trafficking between the endoplasmic reticulum (ER) and the Golgi. All patients had neonatal/early-onset diabetes, severe microcephaly, and epilepsy. YIPF5 is expressed during human brain development, in adult brain and pancreatic islets. We used 3 human ß cell models (YIPF5 silencing in EndoC-ßH1 cells, YIPF5 knockout and mutation knockin in embryonic stem cells, and patient-derived induced pluripotent stem cells) to investigate the mechanism through which YIPF5 loss of function affects ß cells. Loss of YIPF5 function in stem cell-derived islet cells resulted in proinsulin retention in the ER, marked ER stress, and ß cell failure. Partial YIPF5 silencing in EndoC-ßH1 cells and a patient mutation in stem cells increased the ß cell sensitivity to ER stress-induced apoptosis. We report recessive YIPF5 mutations as the genetic cause of a congenital syndrome of microcephaly, epilepsy, and neonatal/early-onset diabetes, highlighting a critical role of YIPF5 in ß cells and neurons. We believe this is the first report of mutations disrupting the ER-to-Golgi trafficking, resulting in diabetes.


Subject(s)
Diabetes Mellitus , Endoplasmic Reticulum Stress/genetics , Genetic Diseases, Inborn , Infant, Newborn, Diseases , Microcephaly , Mutation , Vesicular Transport Proteins , Cell Line , Diabetes Mellitus/embryology , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Female , Genetic Diseases, Inborn/embryology , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Infant, Newborn , Infant, Newborn, Diseases/embryology , Infant, Newborn, Diseases/genetics , Infant, Newborn, Diseases/pathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Microcephaly/embryology , Microcephaly/genetics , Microcephaly/pathology , Neurons/metabolism , Neurons/pathology , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
8.
Methods Mol Biol ; 2128: 55-67, 2020.
Article in English | MEDLINE | ID: mdl-32180185

ABSTRACT

Diabetes is a major public health problem: it is estimated that 420 million people are affected globally. Monogenic forms of diabetes are less common, but variants in monogenic diabetes genes have been shown to contribute to type 2 diabetes risk. In vitro and in vivo models of monogenic forms of diabetes related to the endoplasmic reticulum (ER) stress response provided compelling evidence on the role of ER stress and dysregulated ER stress signaling on ß cell demise in type 1 and type 2 diabetes. In this chapter, we describe the genetics, background, and phenotype of ER stress-related monogenic diabetes mouse models, and we comment on their advantages and disadvantages. We conclude that these mouse models are very useful tools for monogenic diabetes molecular pathogenesis studies, although there is a variability on the methodology that is used. Regarding the use of these models for therapeutic testing of ER stress modulators, a specific consideration should be given to the fact that they recapitulate some, but not all, the phenotypic characteristics of the human disease.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Glucose Intolerance/physiopathology , Insulin Secretion , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Mutant Strains , Phenotype , Signal Transduction
9.
JCI Insight ; 5(2)2020 01 30.
Article in English | MEDLINE | ID: mdl-31877117

ABSTRACT

Friedreich ataxia is an autosomal recessive neurodegenerative disease associated with a high diabetes prevalence. No treatment is available to prevent or delay disease progression. Friedreich ataxia is caused by intronic GAA trinucleotide repeat expansions in the frataxin-encoding FXN gene that reduce frataxin expression, impair iron-sulfur cluster biogenesis, cause oxidative stress, and result in mitochondrial dysfunction and apoptosis. Here we examined the metabolic, neuroprotective, and frataxin-inducing effects of glucagon-like peptide-1 (GLP-1) analogs in in vivo and in vitro models and in patients with Friedreich ataxia. The GLP-1 analog exenatide improved glucose homeostasis of frataxin-deficient mice through enhanced insulin content and secretion in pancreatic ß cells. Exenatide induced frataxin and iron-sulfur cluster-containing proteins in ß cells and brain and was protective to sensory neurons in dorsal root ganglia. GLP-1 analogs also induced frataxin expression, reduced oxidative stress, and improved mitochondrial function in Friedreich ataxia patients' induced pluripotent stem cell-derived ß cells and sensory neurons. The frataxin-inducing effect of exenatide was confirmed in a pilot trial in Friedreich ataxia patients, showing modest frataxin induction in platelets over a 5-week treatment course. Taken together, GLP-1 analogs improve mitochondrial function in frataxin-deficient cells and induce frataxin expression. Our findings identify incretin receptors as a therapeutic target in Friedreich ataxia.


Subject(s)
Exenatide/pharmacology , Friedreich Ataxia/drug therapy , Gene Expression Regulation/drug effects , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Mitochondria/metabolism , Adolescent , Adult , Aged , Animals , Brain/pathology , Cerebellum/pathology , Disease Models, Animal , Exenatide/therapeutic use , Female , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Ganglia, Spinal/pathology , Gene Knock-In Techniques , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Iron/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Oxidative Stress , Reactive Oxygen Species/metabolism , Trinucleotide Repeat Expansion , Young Adult , Frataxin
10.
Diabetologia ; 63(2): 395-409, 2020 02.
Article in English | MEDLINE | ID: mdl-31796987

ABSTRACT

AIMS/HYPOTHESIS: During the onset of type 2 diabetes, excessive dietary intake of saturated NEFA and fructose lead to impaired insulin production and secretion by insulin-producing pancreatic beta cells. The majority of data on the deleterious effects of lipids on functional beta cell mass were obtained either in vivo in rodent models or in vitro using rodent islets and beta cell lines. Translating data from rodent to human beta cells remains challenging. Here, we used the human beta cell line EndoC-ßH1 and analysed its sensitivity to a lipotoxic and glucolipotoxic (high palmitate with or without high glucose) insult, as a way to model human beta cells in a type 2 diabetes environment. METHODS: EndoC-ßH1 cells were exposed to palmitate after knockdown of genes related to saturated NEFA metabolism. We analysed whether and how palmitate induces apoptosis, stress and inflammation and modulates beta cell identity. RESULTS: EndoC-ßH1 cells were insensitive to the deleterious effects of saturated NEFA (palmitate and stearate) unless stearoyl CoA desaturase (SCD) was silenced. SCD was abundantly expressed in EndoC-ßH1 cells, as well as in human islets and human induced pluripotent stem cell-derived beta cells. SCD silencing induced markers of inflammation and endoplasmic reticulum stress and also IAPP mRNA. Treatment with the SCD products oleate or palmitoleate reversed inflammation and endoplasmic reticulum stress. Upon SCD knockdown, palmitate induced expression of dedifferentiation markers such as SOX9, MYC and HES1. Interestingly, SCD knockdown by itself disrupted beta cell identity with a decrease in mature beta cell markers INS, MAFA and SLC30A8 and decreased insulin content and glucose-stimulated insulin secretion. CONCLUSIONS/INTERPRETATION: The present study delineates an important role for SCD in the protection against lipotoxicity and in the maintenance of human beta cell identity. DATA AVAILABILITY: Microarray data and all experimental details that support the findings of this study have been deposited in in the GEO database with the GSE130208 accession code.


Subject(s)
Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Palmitic Acid/pharmacology , Stearoyl-CoA Desaturase/metabolism , Apoptosis/drug effects , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Insulin Secretion/drug effects , Proto-Oncogene Proteins c-myc/metabolism , SOX9 Transcription Factor/metabolism , Transcription Factor HES-1/metabolism
11.
Endocrinology ; 160(5): 1262-1274, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30907926

ABSTRACT

tRNAs are crucial noncoding RNA molecules that serve as amino acid carriers during protein synthesis. The transcription of tRNA genes is a highly regulated process. The tRNA pool is tissue and cell specific, it varies during development, and it is modulated by the environment. tRNAs are highly posttranscriptionally modified by specific tRNA-modifying enzymes. The tRNA modification signature of a cell determines the tRNA epitranscriptome. Perturbations in the tRNA epitranscriptome, as a consequence of mutations in tRNAs and tRNA-modifying enzymes or environmental exposure, have been associated with human disease, including diabetes. tRNA fragmentation induced by impaired tRNA modifications or dietary factors has been linked to pancreatic ß-cell demise and paternal inheritance of metabolic traits. Herein, we review recent findings that associate tRNA epitranscriptome perturbations with diabetes.


Subject(s)
Diabetes Mellitus/genetics , Insulin-Secreting Cells/metabolism , Protein Biosynthesis , RNA Processing, Post-Transcriptional , RNA, Transfer/genetics , Animals , Diabetes Mellitus/metabolism , Humans , Models, Genetic , Mutation , RNA, Transfer/metabolism , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism
12.
Ann Clin Transl Neurol ; 6(1): 161-166, 2019 01.
Article in English | MEDLINE | ID: mdl-30656194

ABSTRACT

Diabetes is a common complication of Friedreich ataxia, requiring sensitive diagnostic methods. Here, we compared the performance of different tests that assess glucose tolerance, insulin sensitivity, and ß-cell function in Friedreich ataxia patients, heterozygous FXN mutation carriers and controls. We find that diabetes is underdiagnosed with fasting glucose alone. The oral glucose tolerance test (OGTT) provides 1.2- to 3.5-fold more diagnoses of impaired glucose homeostasis and diabetes, and adequately measures insulin sensitivity, insulin secretion, and ß-cell function. Clinicians in charge of Friedreich ataxia patients and researchers should incorporate the OGTT as an accurate diagnostic and research tool.


Subject(s)
Blood Glucose/metabolism , Diabetes Complications/diagnosis , Friedreich Ataxia/complications , Glucose Tolerance Test , Adult , Diabetes Complications/metabolism , Female , Friedreich Ataxia/metabolism , Humans , Insulin Resistance , Insulin-Secreting Cells/metabolism , Male , Middle Aged
13.
Nucleic Acids Res ; 46(19): 10302-10318, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30247717

ABSTRACT

Transfer RNAs (tRNAs) are non-coding RNA molecules essential for protein synthesis. Post-transcriptionally they are heavily modified to improve their function, folding and stability. Intronic polymorphisms in CDKAL1, a tRNA methylthiotransferase, are associated with increased type 2 diabetes risk. Loss-of-function mutations in TRMT10A, a tRNA methyltransferase, are a monogenic cause of early onset diabetes and microcephaly. Here we confirm the role of TRMT10A as a guanosine 9 tRNA methyltransferase, and identify tRNAGln and tRNAiMeth as two of its targets. Using RNA interference and induced pluripotent stem cell-derived pancreatic ß-like cells from healthy controls and TRMT10A-deficient patients we demonstrate that TRMT10A deficiency induces oxidative stress and triggers the intrinsic pathway of apoptosis in ß-cells. We show that tRNA guanosine 9 hypomethylation leads to tRNAGln fragmentation and that 5'-tRNAGln fragments mediate TRMT10A deficiency-induced ß-cell death. This study unmasks tRNA hypomethylation and fragmentation as a hitherto unknown mechanism of pancreatic ß-cell demise relevant to monogenic and polygenic forms of diabetes.


Subject(s)
DNA Methylation , Diabetes Mellitus/genetics , Insulin-Secreting Cells/metabolism , Methyltransferases/genetics , RNA, Transfer/metabolism , Aged , Animals , Apoptosis/genetics , Cell Death/genetics , Cell Differentiation/genetics , Cells, Cultured , DNA Fragmentation , Diabetes Mellitus/metabolism , Genetic Linkage , Humans , Induced Pluripotent Stem Cells/physiology , Insulin-Secreting Cells/physiology , Methyltransferases/deficiency , Methyltransferases/metabolism , Middle Aged , Mutation , Rats
14.
J Biol Chem ; 292(36): 14977-14988, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28698383

ABSTRACT

Cytokine-induced endoplasmic reticulum (ER) stress is one of the molecular mechanisms underlying pancreatic ß-cell demise in type 1 diabetes. Thrombospondin 1 (THBS1) was recently shown to promote ß-cell survival during lipotoxic stress. Here we show that ER-localized THBS1 is cytoprotective to rat, mouse, and human ß-cells exposed to cytokines or thapsigargin-induced ER stress. THBS1 confers cytoprotection by maintaining expression of mesencephalic astrocyte-derived neutrotrophic factor (MANF) in ß-cells and thereby prevents the BH3-only protein BIM (BCL2-interacting mediator of cell death)-dependent triggering of the mitochondrial pathway of apoptosis. Prolonged exposure of ß-cells to cytokines or thapsigargin leads to THBS1 and MANF degradation and loss of this prosurvival mechanism. Approaches that sustain intracellular THBS1 and MANF expression in ß-cells should be explored as a cytoprotective strategy in type 1 diabetes.


Subject(s)
Inflammation/metabolism , Insulin-Secreting Cells/metabolism , Nerve Growth Factors/metabolism , Thrombospondin 1/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Endoplasmic Reticulum/metabolism , Humans , Insulin-Secreting Cells/drug effects , Mice , Nerve Growth Factors/antagonists & inhibitors , Oxidative Stress , Thapsigargin/pharmacology
15.
Environ Pollut ; 209: 87-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26647171

ABSTRACT

BACKGROUND: Exposure to particulate matter (PM) is associated with various health effects. Physico-chemical properties influence the toxicological impact of PM, nonetheless the mechanisms underlying PM-induced effects are not completely understood. OBJECTIVES: Human bronchial epithelial cells were used to analyse the pathways activated after exposure to summer and winter urban PM and to identify possible markers of exposure. METHODS: BEAS-2B cells were exposed for 24 h to 10 µg/cm(2) of winter PM2.5 (wPM) and summer PM10 (sPM) sampled in Milan. A microarray technology was used to profile the cells gene expression. Genes and microRNAs were analyzed by bioinformatics technique to identify pathways involved in cellular responses. Selected genes and pathways were validated at protein level (western blot, membrane protein arrays and ELISA). RESULTS: The molecular networks activated by the two PM evidenced a correlation among oxidative stress, inflammation and DNA damage responses. sPM induced the release of pro-inflammatory mediators, although miR-146a and genes related to inflammation resulted up-regulated by both PM. Moreover both PM affected a set of genes, proteins and miRNAs related to antioxidant responses, cancer development, extracellular matrix remodeling and cytoskeleton organization, while miR-29c, implicated in epigenetic modification, resulted up-regulated only by wPM. sPM effects may be related to biological and inorganic components, while wPM apparently related to the high content of organic compounds. CONCLUSIONS: These results may be helpful for the individuation of biomarkers for PM exposure, linked to the specific PM physico-chemical properties.


Subject(s)
Air Pollutants/toxicity , Epithelial Cells/drug effects , Particulate Matter/toxicity , Proteins/genetics , Transcriptome/drug effects , Air Pollutants/analysis , Cell Line , Epithelial Cells/metabolism , Gene Expression , Gene Expression Profiling , Humans , Oxidative Stress , Particulate Matter/analysis , Proteins/metabolism , Seasons
16.
Cell Physiol Biochem ; 34(3): 966-80, 2014.
Article in English | MEDLINE | ID: mdl-25201006

ABSTRACT

BACKGROUND/AIMS: Iodide efflux from thyroid cells into the follicular lumen is essential for the synthesis of thyroid hormones, however, the pathways mediating this transport have only been partially identified. A calcium-activated pathway of iodide efflux has long been recognized, but its molecular identity unknown. Anoctamin 1 (ANO1) is a calcium-activated chloride channel (CaCC), and this study aims to investigate its contribution to iodide fluxes in thyroid cells. METHODS: RT-PCR, immunohistochemistry, and live cell imaging with the fluorescent halide biosensor YFP-H148Q/I152L were used to study the expression, localization and function of ANO1 in thyroid cells. RESULTS: ANO1 mRNA was detected in human thyroid tissue and FRTL-5 thyrocytes, and ANO1 protein was localized to the apical membrane of follicular cells. ATP induced a transient loss of iodide from FRTL-5 cells that was dependent on the mobilization of intracellular calcium, and was inhibited by CaCC/ANO1 inhibitors and siRNA against ANO1. Calcium-activated iodide efflux was also observed in CHO cells over-expressing the Sodium Iodide Symporter (NIS) and ANO1. CONCLUSION: ANO1 in thyrocytes functions as a calcium-activated channel mediating iodide efflux, and may contribute to the rapid delivery of iodide into the follicular lumen for the synthesis of thyroid hormones following activation by calcium-mobilizing stimuli.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium/metabolism , Chloride Channels/metabolism , Iodides/metabolism , Thyroid Gland/metabolism , Animals , Anoctamin-1 , Cell Line , Chloride Channels/genetics , Ion Transport , RNA, Messenger/genetics , Rats , Thyroid Gland/cytology
17.
J Allergy Clin Immunol ; 112(4 Suppl): S78-82, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14530792

ABSTRACT

Intercellular adhesion molecules (ICAMs), in particular ICAM-1, appear to play a crucial role in the recruitment and migration of inflammatory cells to the site of an allergic reaction. Glucocorticoids and allergen-specific immunotherapy have been shown to exert effects on selected components of this system, both in vitro and in vivo, but further research is required to better understand the effects of these therapies. Nasal and conjunctival challenge models (including natural and experimental allergen exposure) represent useful and safe tools for studying the activity of antiallergy drugs in vivo. These tests allow the investigation of a wide variety of parameters including inflammatory infiltrate, ICAM-1 expression, and changes in the concentration of soluble inflammatory mediators. With these tools, anti-inflammatory activity related to the modulation of epithelial cell adhesion molecules has been demonstrated in vivo for several H(1)-receptor antagonists (azelastine, cetirizine, loratadine, levocabastine, oxatomide, and terfenadine). Fexofenadine is a nonsedating, long-acting antihistamine with highly selective H(1)-receptor antagonist activity and a particularly favorable safety profile. In addition, fexofenadine has proven anti-inflammatory activity and has been shown to inhibit a number of mediators at clinically relevant concentrations, including in vitro inhibition of ICAM-1 expression on conjunctival and nasal epithelial cells.


Subject(s)
Anti-Allergic Agents/therapeutic use , Histamine H1 Antagonists/therapeutic use , Hypersensitivity/drug therapy , Terfenadine/analogs & derivatives , Terfenadine/therapeutic use , Anti-Allergic Agents/immunology , Cell Adhesion Molecules/classification , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/physiology , Histamine H1 Antagonists/immunology , Humans , Hypersensitivity/immunology , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Terfenadine/immunology , Treatment Outcome
18.
Ann Allergy Asthma Immunol ; 91(1): 71-8, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12877453

ABSTRACT

BACKGROUND: Chronic rhinosinusitis is frequently associated with asthma. A Th2 cytokine pattern has been recently reported in chronic rhinosinusitis in asthmatic children. OBJECTIVE: To evaluate the effects of treating concomitant chronic rhinosinusitis on respiratory symptoms and function and immunopathological parameters in asthmatic children. METHODS: Eighteen children with moderate asthma (age range, 5 to 12 years) poorly controlled by high doses of inhaled corticosteroids and chronic rhinosinusitis were evaluated for symptoms, spirometry, and inflammation at baseline, after treatment, and 1 month after suspension of treatment. All of the children were treated with a combination of amoxicillin and clavulanate (20 mg/kg twice daily) and fluticasone propionate aqueous nasal spray (100 microg/d) for 14 days. A short course of oral corticosteroids was also prescribed (deflazacort, 1 mg/kg daily for 2 days, 0.5 mg/kg daily for 4 days, and 0.25 mg/kg daily for 4 days). Rhinosinusal lavage for cytokine measurements and a nasal scraping for cytologic analysis were performed in all patients before and after medical treatment. RESULTS: A negative endoscopy result was demonstrated in 15 children after treatment. Symptoms and respiratory function significantly improved after treatment and 1 month later; 8 children had intermittent asthma and 10 had mild asthma. A significant reduction of inflammatory cell numbers was detected in all asthmatic children. Interleukin 4 levels significantly decreased (P < 0.001), whereas interferon-y levels increased (P < 0.001). CONCLUSION: Treatment of chronic rhinosinusitis is able to improve symptoms and respiratory function in asthmatic children, reducing inflammatory cells and reversing the cytokine pattern from a Th2 toward a Th1 profile.


Subject(s)
Amoxicillin-Potassium Clavulanate Combination/therapeutic use , Androstadienes/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Asthma/complications , Drug Therapy, Combination/therapeutic use , Rhinitis, Allergic, Perennial/complications , Sinusitis/complications , Asthma/drug therapy , Child , Child, Preschool , Endoscopy , Female , Fluticasone , Forced Expiratory Volume , Humans , Interferon-gamma/metabolism , Interleukin-4/metabolism , Male , Neutrophils/metabolism , Pregnenediones/therapeutic use , Rhinitis, Allergic, Perennial/drug therapy , Sinusitis/drug therapy
19.
Pediatr Allergy Immunol ; 14(3): 238-41, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12787306

ABSTRACT

A Th2 cytokine pattern has recently been reported both in allergic and nonallergic chronic rhinosinusitis in asthmatic children. The aim of the study was to evaluate the cytokine pattern in chronic rhinosinusitis in allergic and nonallergic asthmatic children before and after medical treatment. Thirty asthmatic children were evaluated, 18 males and 12 females (mean age 9.1 years). Sixteen were allergic and 14 were nonallergic. All children were asthmatic and suffered from chronic rhinosinusitis, whose diagnosis was confirmed by endoscopy. All of them were treated with amoxicilline-clavulanate (20 mg/kg b.i.d.) and fluticasone propionate aqueous nasal spray (100 microg daily) for 14 days; a short course of oral corticosteroid was also prescribed (deflazacort 1 mg/kg daily for 2 days, 0.5 mg/kg daily for 4 days and 0.25 mg/kg daily for 4 days). Rhinosinusal lavage and nasal cytology were performed in all subjects before and after medical treatment. IL4 and IFNgamma were measured by immunoassay and inflammatory cells were counted by conventional staining. Thirteen allergic children and 12 nonallergic children showed a negative endoscopy after the treatment. Allergic subjects showed a significant decrease of IL4 (p = 0.0002) and a significant increase of IFNgamma (p = 0.03) after the treatment. Nonallergic children showed a significant decrease of IL4 (p = 0.0007) and a nonsignificant increase of IFNgamma. A significant reduction of the inflammatory infiltrate was detected in all asthmatic children (p < 0.05). This study confirms a Th2 polarization in chronic rhinosinusitis both in allergic and nonallergic asthmatic children. Moreover, the medical treatment of chronic rhinosinusitis reversed the cytokine pattern from a Th2 towards a Th1 profile both in allergic and nonallergic children.


Subject(s)
Amoxicillin-Potassium Clavulanate Combination/therapeutic use , Androstadienes/therapeutic use , Anti-Allergic Agents/therapeutic use , Anti-Bacterial Agents/therapeutic use , Asthma/drug therapy , Interferon-gamma/drug effects , Interleukin-4/metabolism , Sinusitis/drug therapy , Amoxicillin-Potassium Clavulanate Combination/pharmacology , Androstadienes/pharmacology , Anti-Allergic Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Asthma/complications , Asthma/immunology , Child , Child, Preschool , Chronic Disease , Endoscopy , Female , Fluticasone , Humans , Interferon-gamma/metabolism , Male , Sinusitis/complications , Sinusitis/immunology , Th2 Cells/drug effects , Th2 Cells/metabolism , Treatment Outcome
20.
Ann Allergy Asthma Immunol ; 90(4): 434-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12722967

ABSTRACT

BACKGROUND: Some second-generation antihistamines have anti-inflammatory activities, but the clinical relevance of this property is still unclear. OBJECTIVE: The aim of our study was to investigate the effects of azelastine when administered during the early-phase reaction. METHODS: This investigation was designed as a randomized, placebo-controlled, double-blind, parallel-group study. Clinical and inflammatory events were evaluated after a single dose of azelastine eyedrops or placebo was administered 30 minutes after an allergen-specific conjunctival challenge. Twenty outpatients with allergic rhinoconjunctivitis attributable to Parietaria judaica were enrolled in the study outside the pollen season. Patients were evaluated at baseline, after allergen challenge (at 30 minutes), and after administration of azelastine (at 30 minutes and at 6 hours). The following variables were evaluated: hyperemia, lacrimation, itching, eyelid swelling, number of inflammatory cells (neutrophils, eosinophils, monocytes, and lymphocytes), and intercellular adhesion molecule-1 expression on conjunctival epithelial cells. RESULTS: Azelastine, in comparison to placebo, significantly reduced symptom scores, number of inflammatory cells, and intercellular adhesion molecule-1 expression during the early- and late-phase reaction. CONCLUSIONS: The ability of azelastine to reduce symptoms and inflammation during an ongoing allergic reaction can be considered concrete and convincing proof of a clinically relevant anti-inflammatory activity.


Subject(s)
Anti-Allergic Agents/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Conjunctivitis, Allergic/drug therapy , Phthalazines/therapeutic use , Adolescent , Adult , Allergens/adverse effects , Anti-Allergic Agents/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cell Count , Chemotaxis, Leukocyte/drug effects , Conjunctiva/metabolism , Conjunctiva/pathology , Conjunctivitis, Allergic/complications , Double-Blind Method , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation/drug effects , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Male , Middle Aged , NF-kappa B/metabolism , Ophthalmic Solutions , Parietaria , Phthalazines/administration & dosage , Pollen/adverse effects , Radioallergosorbent Test , Rhinitis, Allergic, Seasonal/complications , Skin Tests , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...