Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hepatology ; 75(2): 309-321, 2022 02.
Article in English | MEDLINE | ID: mdl-34766362

ABSTRACT

BACKGROUND AND AIMS: Gallbladder cancer (GBC) has a female predominance, whereas the other biliary tract cancers (BTCs) have a male predominance, suggesting that sex hormones may be involved in carcinogenesis. We sought to evaluate the association between menopausal hormone therapy (MHT) and the risk of BTC in women. APPROACH AND RESULTS: This nested case-control study was conducted in the UK Clinical Practice Research Datalink. Cases diagnosed between 1990 and 2017 with incident primary cancers of the gallbladder (GBC), cholangiocarcinoma (CCA), ampulla of Vater (AVC), and mixed type were matched to 5 controls on birth year, diagnosis year, and years in the general practice using incidence density sampling. Conditional logistic regression was used to calculate ORs and 95% CIs for associations between MHT use and BTC type. The sample consisted of 1,682 BTC cases (483 GBC, 870 CCA, 105 AVC, and 224 mixed) and 8,419 matched controls with a mean age of 73 (SD, 11) years. Combined formulations (estrogen-progesterone) were associated with an increased GBC risk (OR, 1.97; 95% CI, 1.08, 3.59). Orally administered MHT was associated with an increased GBC risk (OR, 2.28; 95% CI, 1.24, 4.17). Estrogen-only formulations (OR, 0.59; 95% CI, 0.34, 0.93) and cream or suppository administrations (OR, 0.57; 95% CI, 0.34, 0.95) were associated with decreased CCA risk. The number of prescriptions, dose, duration of use, and time since last use were not associated with GBC or CCA risk. MHT use was not associated with risk of AVC or mixed cancer. CONCLUSIONS: Combination MHT formulations and oral administrations were associated with increased GBC risk, whereas estrogen-only formulations were associated with a lower CCA risk. MHT formulation and administration should be carefully considered when prescribing.


Subject(s)
Ampulla of Vater , Cholangiocarcinoma/epidemiology , Common Bile Duct Neoplasms/epidemiology , Gallbladder Neoplasms/epidemiology , Hormone Replacement Therapy , Neoplasms, Complex and Mixed/epidemiology , Administration, Oral , Administration, Topical , Aged , Aged, 80 and over , Case-Control Studies , Drug Combinations , Estrogens/therapeutic use , Female , Humans , Incidence , Menopause , Middle Aged , Progesterone/therapeutic use , Progestins/therapeutic use , Risk Factors , Suppositories , United Kingdom/epidemiology
2.
Hum Genet ; 141(1): 147-173, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34889978

ABSTRACT

The combined impact of common and rare exonic variants in COVID-19 host genetics is currently insufficiently understood. Here, common and rare variants from whole-exome sequencing data of about 4000 SARS-CoV-2-positive individuals were used to define an interpretable machine-learning model for predicting COVID-19 severity. First, variants were converted into separate sets of Boolean features, depending on the absence or the presence of variants in each gene. An ensemble of LASSO logistic regression models was used to identify the most informative Boolean features with respect to the genetic bases of severity. The Boolean features selected by these logistic models were combined into an Integrated PolyGenic Score that offers a synthetic and interpretable index for describing the contribution of host genetics in COVID-19 severity, as demonstrated through testing in several independent cohorts. Selected features belong to ultra-rare, rare, low-frequency, and common variants, including those in linkage disequilibrium with known GWAS loci. Noteworthily, around one quarter of the selected genes are sex-specific. Pathway analysis of the selected genes associated with COVID-19 severity reflected the multi-organ nature of the disease. The proposed model might provide useful information for developing diagnostics and therapeutics, while also being able to guide bedside disease management.


Subject(s)
COVID-19/genetics , COVID-19/physiopathology , Exome Sequencing , Genetic Predisposition to Disease , Phenotype , Severity of Illness Index , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Germany , Humans , Italy , Male , Middle Aged , Polymorphism, Single Nucleotide , Quebec , SARS-CoV-2 , Sweden , United Kingdom
3.
Proc Natl Acad Sci U S A ; 116(33): 16507-16512, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31371497

ABSTRACT

The retina is an extension of the brain. Like the brain, neurodegeneration of the retina occurs with age and is the cause of several retinal diseases including optic neuritis, macular degeneration, and glaucoma. Liver X receptors (LXRs) are expressed in the brain where they play a key role in maintenance of cerebrospinal fluid and the health of dopaminergic neurons. Herein, we report that LXRs are expressed in the retina and optic nerve and that loss of LXRß, but not LXRα, leads to loss of ganglion cells in the retina. In the retina of LXRß-/- mice, there is an increase in amyloid A4 and deposition of beta-amyloid (Aß) aggregates but no change in the level of apoptosis or autophagy in the ganglion cells and no activation of microglia or astrocytes. However, in the optic nerve there is a loss of aquaporin 4 (AQP4) in astrocytes and an increase in activation of microglia. Since loss of AQP4 and microglial activation in the optic nerve precedes the loss of ganglion cells, and accumulation of Aß in the retina, the cause of the neuronal loss appears to be optic nerve degeneration. In patients with optic neuritis there are frequently AQP4 autoantibodies which block the function of AQP4. LXRß-/- mouse is another model of optic neuritis in which AQP4 antibodies are not detectable, but AQP4 function is lost because of reduction in its expression.


Subject(s)
Liver X Receptors/deficiency , Nerve Degeneration/pathology , Optic Nerve/pathology , Retina/pathology , Amyloid beta-Peptides/metabolism , Animals , Aquaporin 4/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Female , Liver X Receptors/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Oligodendroglia/metabolism , Optic Nerve/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
4.
Am J Physiol Gastrointest Liver Physiol ; 321(4): G243-G251, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34259574

ABSTRACT

The gallbladder is considered an important organ in maintaining digestive and metabolic homeostasis. Given that therapeutic options for gallbladder diseases are often limited to cholecystectomy, understanding gallbladder pathophysiology is essential in developing novel therapeutic strategies. Since liver X receptor ß (LXRß), an oxysterol-activated transcription factor, is strongly expressed in gallbladder cholangiocytes, the aim was to investigate LXRß physiological function in the gallbladder. Thus, we studied the gallbladders of WT and LXRß-/- male mice using immunohistochemistry, electron microscopy, qRT-PCR, bile duct cannulation, bile and blood biochemistry, and duodenal pH measurements. LXRß-/- mice presented a large gallbladder bile volume with high duodenal mRNA levels of the vasoactive intestinal polypeptide (VIP), a strong mediator of gallbladder relaxation. LXRß-/- gallbladders showed low mRNA and protein expression of Aquaporin-1, Aquaporin-8, and cystic fibrosis transmembrane conductance regulator (CFTR). A cystic fibrosis-resembling phenotype was evident in the liver showing high serum cholestatic markers and the presence of reactive cholangiocytes. For LXRß being a transcription factor, we identified eight putative binding sites of LXR on the promoter and enhancer of the Cftr gene, suggesting Cftr as a novel LXRß regulated gene. In conclusion, LXRß was recognized as a regulator of gallbladder bile volume through multiple mechanisms involving CFTR and aquaporins.NEW & NOTEWORTHY This report reveals a novel and specific role of the nuclear receptor liver X receptor ß (LXRß) in controlling biliary tree pathophysiology. LXRß-/- mice have high gallbladder bile volume and are affected by a cholangiopathy that resembles cystic fibrosis. We found LXRß to regulate the expression of both aquaporins water channels and the cystic fibrosis transmembrane conductance regulator. This opens a new field in biliary tree pathophysiology, enlightening a possible transcription factor controlling CFTR expression.


Subject(s)
Aquaporin 1/metabolism , Aquaporins/metabolism , Bile/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gallbladder/metabolism , Liver X Receptors/metabolism , Animals , Aquaporin 1/genetics , Aquaporins/genetics , Binding Sites , Cell Proliferation , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Duodenum/metabolism , Gallbladder/ultrastructure , Liver X Receptors/genetics , Male , Mice, Knockout , Promoter Regions, Genetic , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
5.
N Engl J Med ; 381(14): 1395-1396, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31577894
6.
Proc Natl Acad Sci U S A ; 109(8): 3030-4, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22323586

ABSTRACT

The present study demonstrates a key role for the oxysterol receptor liver X receptor ß (LXRß) in the etiology of diabetes insipidus (DI). Given free access to water, LXRß(-/-) but not LXRα(-/-) mice exhibited polyuria (abnormal daily excretion of highly diluted urine) and polydipsia (increased water intake), both features of diabetes insipidus. LXRß(-/-) mice responded to 24-h dehydration with a decreased urine volume and increased urine osmolality. To determine whether the DI was of central or nephrogenic origin, we examined the responsiveness of the kidney to arginine vasopressin (AVP). An i.p. injection of AVP to LXRß(-/-) mice revealed a partial kidney response: There was no effect on urine volume, but there was a significant increase of urine osmolality, suggesting that DI may be caused by a defect in central production of AVP. In the brain of WT mice LXRß was expressed in the nuclei of magnocellular neurons in the supraoptic and paraventricular nuclei of the hypothalamus. In LXRß(-/-) mice the expression of AVP was markedly decreased in the magnocellular neurons as well as in urine collected over a 24-h period. The persistent high urine volume after AVP administration was traced to a reduction in aquaporin-1 expression in the kidney of LXRß(-/-) mice. The LXR agonist (GW3965) in WT mice elicited an increase in urine osmolality, suggesting that LXRß is a key receptor in controlling water balance with targets in both the brain and kidney, and it could be a therapeutic target in disorders of water balance.


Subject(s)
Aquaporin 1/metabolism , Diabetes Insipidus, Neurogenic/metabolism , Kidney/metabolism , Orphan Nuclear Receptors/deficiency , Animals , Arginine Vasopressin/administration & dosage , Arginine Vasopressin/pharmacology , Arginine Vasopressin/urine , Benzoates/administration & dosage , Benzoates/pharmacology , Benzylamines/administration & dosage , Benzylamines/pharmacology , Body Water , Dehydration/blood , Dehydration/complications , Dehydration/physiopathology , Dehydration/urine , Diabetes Insipidus, Neurogenic/complications , Diabetes Insipidus, Neurogenic/pathology , Diabetes Insipidus, Neurogenic/physiopathology , Female , Kidney/pathology , Kidney/physiopathology , Liver X Receptors , Mice , Neurons/drug effects , Neurons/metabolism , Orphan Nuclear Receptors/metabolism , Osmolar Concentration , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology , Paraventricular Hypothalamic Nucleus/physiopathology , Polydipsia/blood , Polydipsia/complications , Polydipsia/physiopathology , Polydipsia/urine , Polyuria/blood , Polyuria/complications , Polyuria/physiopathology , Polyuria/urine , Supraoptic Nucleus/drug effects , Supraoptic Nucleus/metabolism , Supraoptic Nucleus/pathology , Supraoptic Nucleus/physiopathology , Water-Electrolyte Balance/physiology
7.
Biochem Biophys Res Commun ; 446(3): 647-50, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24300092

ABSTRACT

The two Liver X Receptors, LXRα and LXRß, are nuclear receptors belonging to the superfamily of ligand-activated transcription factors. They share more than 78% homology in amino acid sequence, a common profile of oxysterol ligands and the same heterodimerization partner, Retinoid X Receptor. LXRs play crucial roles in several metabolic pathways: lipid metabolism, in particular in preventing cellular cholesterol accumulation; glucose homeostasis; inflammation; central nervous system functions and water transport. As with all nuclear receptors, the transcriptional activity of LXR is the result of an orchestration of numerous cellular factors including ligand bioavailability, presence of corepressors and coactivators and cellular context i.e., what other pathways are activated in the cell at the time the receptor recognizes its ligand. In this mini-review we summarize the factors regulating the transcriptional activity and the mechanisms of action of these two receptors.


Subject(s)
Hydroxycholesterols/metabolism , Orphan Nuclear Receptors/physiology , Animals , Homeostasis , Humans , Liver X Receptors , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism
8.
Sci Rep ; 14(1): 191, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38168135

ABSTRACT

15% of US adults have gallstones, most of which are clinically "silent". Several studies show that menopausal hormone therapy (MHT) increases symptomatic gallstones and cholecystectomy risk. MHT use may be contraindicated in women with gallstones and population studies may be biased by "confounding by contraindication" while the true association between MHT and gallstones remains underestimated. We sought to examine whether MHT use was associated with asymptomatic gallstones using instrumental variable (IV) analysis to account for confounding by contraindication. We used 2018 postmenopausal women from the Third National Health and Nutrition Examination Survey to estimate associations of MHT use with asymptomatic gallstones. A traditional logistic regression analysis was compared to instrumental variable (IV) analysis to account for confounding by contraindication. 12% of women with asymptomatic gallstones and 25% of women without gallstones were current MHT users (P < 0.001). The traditional analysis suggested a decreased odds of asymptomatic gallstones in current versus never users (OR 0.58, 95% CI 0.37, 0.89), but increased odds (OR 1.51, 95% CI 0.44, 5.16) in the IV analysis. The traditional analysis consistently underestimated the odds of asymptomatic gallstones with MHT use compared to the IV analysis. Accounting for confounding by contraindication, we found a suggestive, though imprecise, positive association between MHT use and asymptomatic gallstones among postmenopausal women. Failure to consider contraindication can produce incorrect results.


Subject(s)
Gallstones , Adult , Female , Humans , Gallstones/epidemiology , Gallstones/etiology , Estrogen Replacement Therapy/adverse effects , Nutrition Surveys , Surveys and Questionnaires , Menopause , Hormone Replacement Therapy
9.
Hepatology ; 56(6): 2288-96, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22729460

ABSTRACT

UNLABELLED: Nuclear receptors (NRs) play crucial roles in the regulation of hepatic cholesterol synthesis, metabolism, and conversion to bile acids, but their actions in cholangiocytes have not been examined. In this study, we investigated the roles of NRs in cholangiocyte physiology and cholesterol metabolism and flux. We examined the expression of NRs and other genes involved in cholesterol homeostasis in freshly isolated and cultured murine cholangiocytes and found that these cells express a specific subset of NRs, including liver X receptor (LXR) ß and peroxisome proliferator-activated receptor (PPAR) δ. Activation of LXRß and/or PPARδ in cholangiocytes induces ATP-binding cassette cholesterol transporter A1 (ABCA1) and increases cholesterol export at the basolateral compartment in polarized cultured cholangiocytes. In addition, PPARδ induces Niemann-Pick C1-like L1 (NPC1L1), which imports cholesterol into cholangiocytes and is expressed on the apical cholangiocyte membrane via specific interaction with a peroxisome proliferator-activated response element (PPRE) within the NPC1L1 promoter. CONCLUSION: We propose that (1) LXRß and PPARδ coordinate NPC1L1/ABCA1-dependent vectorial cholesterol flux from bile through cholangiocytes and (2) manipulation of these processes may influence bile composition with important applications in cholestatic liver disease and gallstone disease, two serious health concerns for humans.


Subject(s)
Cholesterol/metabolism , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , PPAR delta/genetics , PPAR delta/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Bile Ducts/cytology , Bile Ducts/metabolism , Cells, Cultured , Gene Expression Profiling , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , Homeostasis/genetics , Liver X Receptors , Membrane Transport Proteins/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pregnane X Receptor , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Proc Natl Acad Sci U S A ; 107(33): 14763-8, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20679224

ABSTRACT

Gallbladder cancer is a highly aggressive disease with poor prognosis that is two to six times more frequent in women than men. The development of gallbladder cancer occurs over a long time (more than 15 y) and evolves from chronic inflammation to dysplasia/metaplasia, carcinoma in situ, and invasive carcinoma. In the present study we found that, in female mice in which the oxysterol receptor liver X receptor-beta (LXRbeta) has been inactivated, preneoplastic lesions of the gallbladder developed and evolved to cancer in old animals. LXRbeta is a nuclear receptor involved in the control of lipid homeostasis, glucose metabolism, inflammation, proliferation, and CNS development. LXRbeta(-/-) female gallbladders were severely inflamed, with regions of dysplasia and high cell density, hyperchromasia, metaplasia, and adenomas. No abnormalities were evident in male mice, nor in LXRalpha(-/-) or LXRalpha(-/-)beta(-/-) animals of either sex. Interestingly, the elimination of estrogens with ovariectomy prevented development of preneoplastic lesions in LXRbeta(-/-) mice. The etiopathological mechanism seems to involve TGF-beta signaling, as the precancerous lesions were characterized by strong nuclear reactivity of phospho-SMAD-2 and SMAD-4 and loss of E-cadherin expression. Upon ovariectomy, E-cadherin was reexpressed on the cell membranes and immunoreactivity of pSMAD-2 in the nuclei was reduced. These findings suggest that LXRbeta in a complex interplay with estrogens and TGF-beta could play a crucial role in the malignant transformation of the gallbladder epithelium.


Subject(s)
Estrogens/metabolism , Gallbladder Neoplasms/metabolism , Gallbladder/metabolism , Orphan Nuclear Receptors/metabolism , Age Factors , Animals , Apoptosis , Cadherins/metabolism , Cell Proliferation , Female , Gallbladder/pathology , Gallbladder Neoplasms/genetics , Humans , Immunohistochemistry , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors/genetics , Ovariectomy , Phosphorylation , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Smad2 Protein/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/metabolism
11.
Proc Natl Acad Sci U S A ; 107(14): 6453-8, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20308571

ABSTRACT

At 2 years of age, 100% (23/23) of ERbeta(-/-) female mice have developed large pituitary and ovarian tumors. The pituitary tumors are gonadotropin-positive and the ovarian tumors are sex cord (less differentiated) and granulosa cell tumors (differentiated and estrogen secreting). No male mice had pituitary tumors and no pituitary or ovarian tumors developed in ERalpha(-/-) mice or in ERalphabeta(-/-) double knockout mice. The tumors have high proliferation indices, are ERalpha-positive, ERbeta-negative, and express high levels of nuclear phospho-SMAD3. Mice with granulosa cell tumors also had hyperproliferative endometria. The cause of the pituitary tumors appeared to be excessive secretion of gonadotropin releasing hormone (GnRH) from the hypothalamus resulting from high expression of NPY. The ovarian phenotype is similar to that seen in mice where inhibin is ablated. The data indicate that ERbeta plays an important role in regulating GnRH secretion. We suggest that in the absence of ERbeta, the proliferative action of FSH/SMAD3 is unopposed and the high proliferation leads to the development of ovarian tumors. The absence of tumors in the ERalphabeta(-/-) mice suggests that tumor development requires the presence of ERalpha.


Subject(s)
Aging , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Gonadotropins/metabolism , Neoplasms, Second Primary/metabolism , Ovarian Neoplasms/metabolism , Pituitary Neoplasms/metabolism , Animals , Cell Proliferation , Estrogen Receptor alpha/deficiency , Estrogen Receptor beta/deficiency , Female , Gonadotropin-Releasing Hormone/metabolism , Ki-67 Antigen/metabolism , Male , Mice , Mice, Knockout , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/pathology , Neuropeptide Y/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Sex Characteristics
14.
J Infect ; 85(3): 318-321, 2022 09.
Article in English | MEDLINE | ID: mdl-35700866

ABSTRACT

COVID-19 has shown a relevant heterogeneity in spread and fatality among countries together with a significant variability in its clinical presentation, indicating that host genetic factors may influence COVID-19 pathogenicity. Indeed, subjects carrying single pathogenic variants of the Cystic Fibrosis (CF) Transmembrane Conductance Regulator (CFTR) gene - i.e. CF carriers - are more susceptible to respiratory tract infections and are more likely to undergo severe COVID-19 with higher risk of 14-day mortality. Given that CF carrier prevalence varies among ethnicities and nations, an ecological study in 37 countries was conducted, in order to determine to what extent the diverse CF carrier geographical distribution may have affected COVID-19 spread and fatality during the first pandemic wave. The CF prevalence in countries, as indicator of the geographical distribution of CF carriers, significantly correlated in a direct manner with both COVID-19 prevalence and its Case Fatality Rate (CFR). In a regression study weighted for the number of tests performed, COVID-19 prevalence positively correlated with CF prevalence, while CFR correlated with population percentage older than 65-year, cancer and CF prevalence. Multivariate regression model also confirmed COVID-19 CFR to be associated with CF prevalence, after adjusting for elderly, cancer prevalence, and weighting for the number of tests performed. This study suggests a putative contribution of population genetics of CFTR in understanding the spatial distribution of COVID-19 spread and fatality.


Subject(s)
COVID-19 , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Heterozygote , COVID-19/epidemiology , COVID-19/mortality , Cystic Fibrosis/complications , Cystic Fibrosis/epidemiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Genetics, Population , Humans , Mutation
15.
Cells ; 11(24)2022 12 16.
Article in English | MEDLINE | ID: mdl-36552859

ABSTRACT

Carriers of single pathogenic variants of the CFTR (cystic fibrosis transmembrane conductance regulator) gene have a higher risk of severe COVID-19 and 14-day death. The machine learning post-Mendelian model pinpointed CFTR as a bidirectional modulator of COVID-19 outcomes. Here, we demonstrate that the rare complex allele [G576V;R668C] is associated with a milder disease via a gain-of-function mechanism. Conversely, CFTR ultra-rare alleles with reduced function are associated with disease severity either alone (dominant disorder) or with another hypomorphic allele in the second chromosome (recessive disorder) with a global residual CFTR activity between 50 to 91%. Furthermore, we characterized novel CFTR complex alleles, including [A238V;F508del], [R74W;D1270N;V201M], [I1027T;F508del], [I506V;D1168G], and simple alleles, including R347C, F1052V, Y625N, I328V, K68E, A309D, A252T, G542*, V562I, R1066H, I506V, I807M, which lead to a reduced CFTR function and thus, to more severe COVID-19. In conclusion, CFTR genetic analysis is an important tool in identifying patients at risk of severe COVID-19.


Subject(s)
COVID-19 , Cystic Fibrosis , Humans , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Alleles , Cystic Fibrosis/pathology , COVID-19/genetics , Heterozygote
16.
Autophagy ; 18(7): 1662-1672, 2022 07.
Article in English | MEDLINE | ID: mdl-34964709

ABSTRACT

The polymorphism L412F in TLR3 has been associated with several infectious diseases. However, the mechanism underlying this association is still unexplored. Here, we show that the L412F polymorphism in TLR3 is a marker of severity in COVID-19. This association increases in the sub-cohort of males. Impaired macroautophagy/autophagy and reduced TNF/TNFα production was demonstrated in HEK293 cells transfected with TLR3L412F-encoding plasmid and stimulated with specific agonist poly(I:C). A statistically significant reduced survival at 28 days was shown in L412F COVID-19 patients treated with the autophagy-inhibitor hydroxychloroquine (p = 0.038). An increased frequency of autoimmune disorders such as co-morbidity was found in L412F COVID-19 males with specific class II HLA haplotypes prone to autoantigen presentation. Our analyses indicate that L412F polymorphism makes males at risk of severe COVID-19 and provides a rationale for reinterpreting clinical trials considering autophagy pathways.Abbreviations: AP: autophagosome; AUC: area under the curve; BafA1: bafilomycin A1; COVID-19: coronavirus disease-2019; HCQ: hydroxychloroquine; RAP: rapamycin; ROC: receiver operating characteristic; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2; TLR: toll like receptor; TNF/TNF-α: tumor necrosis factor.


Subject(s)
COVID-19 , Toll-Like Receptor 3 , Autophagy/genetics , Biomarkers , COVID-19/genetics , HEK293 Cells , Humans , Hydroxychloroquine/therapeutic use , Male , Polymorphism, Single Nucleotide , SARS-CoV-2/genetics , Severity of Illness Index , Toll-Like Receptor 3/genetics
17.
Am J Physiol Endocrinol Metab ; 301(1): E210-22, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21521718

ABSTRACT

The liver X receptors (LXRs) play a key role in cholesterol and bile acid metabolism but are also important regulators of glucose metabolism. Recently, LXRs have been proposed as a glucose sensor affecting LXR-dependent gene expression. We challenged wild-type (WT) and LXRαß(-/-) mice with a normal diet (ND) or a high-carbohydrate diet (HCD). Magnetic resonance imaging showed different fat distribution between WT and LXRαß(-/-) mice. Surprisingly, gonadal (GL) adipocyte volume decreased on HCD compared with ND in WT mice, whereas it slightly increased in LXRαß(-/-) mice. Interestingly, insulin-stimulated lipogenesis of isolated GL fat cells was reduced on HCD compared with ND in LXRαß(-/-) mice, whereas no changes were observed in WT mice. Net de novo lipogenesis (DNL) calculated from Vo(2) and Vco(2) was significantly higher in LXRαß(-/-) than in WT mice on HCD. Histology of HCD-fed livers showed hepatic steatosis in WT mice but not in LXRαß(-/-) mice. Glucose tolerance was not different between groups, but insulin sensitivity was decreased by the HCD in WT but not in LXRαß(-/-) mice. Finally, gene expression analysis of adipose tissue showed induced expression of genes involved in DNL in LXRαß(-/-) mice compared with WT animals as opposed to the liver, where expression of DNL genes was repressed in LXRαß(-/-) mice. We thus conclude that absence of LXRs stimulates DNL in adipose tissue, but suppresses DNL in the liver, demonstrating opposite roles of LXR in DNL regulation in these two tissues. These results show tissue-specific regulation of LXR activity, a crucial finding for drug development.


Subject(s)
Lipogenesis/genetics , Orphan Nuclear Receptors/physiology , Adipocytes/cytology , Adipocytes/metabolism , Adipocytes/physiology , Adipose Tissue/metabolism , Adiposity/genetics , Animals , Body Fat Distribution , Cells, Cultured , Female , Lipolysis/genetics , Lipolysis/physiology , Liver X Receptors , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity/genetics , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism
18.
Proc Natl Acad Sci U S A ; 105(6): 2094-9, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18238900

ABSTRACT

Administration of beta-sitosterol (42 mg/kg per day) for 3 weeks to 8-month-old male LXRbeta-/- mice resulted in the death of motor neurons in the lumbar region of the spinal cord and loss of tyrosine hydroxylase-positive dopaminergic neurons in the substantia nigra. In mice at 5 months of age, beta-sitosterol had no observed toxicity but at 16 months of age, it caused severe paralysis and symptoms typical of dopaminergic dysfunction in LXRbeta-/- mice. WT mice were not affected by these doses of beta-sitosterol. In 5-month-old mice, levels of the intestinal transporters, ABCG5/8 and Niemann-Pick C1 Like 1, were not affected by loss of liver X receptor (LXR) beta and/or treatment with beta-sitosterol nor were there changes in plasma levels of cholesterol or beta-sitosterol. In 8-month-old LXRbeta-/- mice there was activation of microglia in the substantia nigra pars reticulata and aggregates of ubiquitin and TDP-43 in the cytoplasm of large motor neurons in the lumbar spinal cord. Brain cholesterol concentrations were higher in LXRbeta-/- than in their WT counterparts, and treatment with beta-sitosterol reduced brain cholesterol in both WT and LXRbeta-/- mice. In LXRbeta-/- mice but not in WT mice levels of 24-hydrocholesterol were increased upon beta-sitosterol treatment. These data indicate that multiple mechanisms are involved in the sensitivity of LXRbeta-/- mice to beta-sitosterol. These include activation of microglia, accumulation of protein aggregates in the cytoplasm of large motor neurons, and depletion of brain cholesterol.


Subject(s)
Amyotrophic Lateral Sclerosis/chemically induced , DNA-Binding Proteins/physiology , Parkinsonian Disorders/etiology , Receptors, Cytoplasmic and Nuclear/physiology , Sitosterols/toxicity , ATP-Binding Cassette Transporters/metabolism , Animals , Cholesterol/blood , DNA-Binding Proteins/genetics , Dopamine/metabolism , Immunohistochemistry , Intestinal Mucosa/metabolism , Liver X Receptors , Male , Mice , Mice, Knockout , Motor Neurons/drug effects , Motor Neurons/enzymology , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/genetics , Sitosterols/blood , Spinal Cord/drug effects , Spinal Cord/pathology , Substantia Nigra/drug effects , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
19.
Proc Natl Acad Sci U S A ; 105(39): 15052-7, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18806227

ABSTRACT

Liver X receptors (LXRs) alpha and beta are nuclear oxysterol receptors with a key role in cholesterol, triglyceride, and glucose metabolism. In LXRbeta(-/-) mice on a normal diet, there is a reduction in size of perigonadal fat pad and, on high-fat diet there is resistance to obesity. In the present study, we investigated the reason for the resistance of LXRbeta(-/-) mice to weight gain. In LXRbeta(-/-) mice we found pancreatic exocrine insufficiency with reduced serum levels of amylase and lipase, reduced proteolytic activity in feces, chronic inflammatory infiltration, and, in the ductal epithelium, an increased apoptosis without compensatory proliferation. Electron microscopy revealed ductal dilatation with intraductal laminar structures characteristic of cystic fibrosis. To investigate the relationship between LXRbeta and pancreatic secretion, we studied the expression of LXRbeta and the water channel, aquaporin-1 (AQP1), in the ductal epithelium of the pancreas. In WT mice, ductal epithelial cells expressed LXRbeta in the nuclei and AQP1 on the plasma membrane. In LXRbeta(-/-) mice neither LXRbeta nor AQP1 was detectable. Moreover, in WT mice the LXR agonist (T2320) increased AQP1 gene expression. These data demonstrate that in LXRbeta(-/-) mice dietary resistance to weight gain is caused by pancreatic insufficiency and that LXRbeta regulates pancreatic exocrine secretion through the control of AQP1 expression. Pancreatic exocrine insufficiency is the main cause of malabsorption syndrome responsible for weight loss in adults and growth failure in children. Several genes are known to be involved in the pathogenesis and susceptibility to pancreatic insufficiency. LXRbeta should be included in that list.


Subject(s)
Aquaporin 1/biosynthesis , DNA-Binding Proteins/genetics , Exocrine Pancreatic Insufficiency/genetics , Exocrine Pancreatic Insufficiency/metabolism , Pancreas, Exocrine/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Weight Gain/genetics , Adipose Tissue/metabolism , Animals , Aquaporin 1/genetics , Body Weight/genetics , DNA-Binding Proteins/agonists , Exocrine Pancreatic Insufficiency/pathology , Female , Gene Expression Regulation/drug effects , Liver X Receptors , Male , Mice , Mice, Knockout , Orphan Nuclear Receptors , Pancreas, Exocrine/ultrastructure , Pancreatic Ducts/metabolism , Pancreatic Ducts/ultrastructure , RNA, Messenger/biosynthesis , Receptors, Cytoplasmic and Nuclear/agonists
20.
J Pers Med ; 11(6)2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34203982

ABSTRACT

The clinical presentation of COVID-19 is extremely heterogeneous, ranging from asymptomatic to severely ill patients. Thus, host genetic factors may be involved in determining disease presentation and progression. Given that carriers of single cystic fibrosis (CF)-causing variants of the CFTR gene-CF-carriers-are more susceptible to respiratory tract infections, our aim was to determine their likelihood of undergoing severe COVID-19. We implemented a cohort study of 874 individuals diagnosed with COVID-19, during the first pandemic wave in Italy. Whole exome sequencing was performed and validated CF-causing variants were identified. Forty subjects (16 females and 24 males) were found to be CF-carriers. Among mechanically ventilated patients, CF-carriers were more represented (8.7%) and they were significantly (p < 0.05) younger (mean age 51 years) compared to noncarriers (mean age 61.42 years). Furthermore, in the whole cohort, the age of male CF-carriers was lower, compared to noncarriers (p < 0.05). CF-carriers had a relative risk of presenting an abnormal inflammatory response (CRP ≥ 20 mg/dL) of 1.69 (p < 0.05) and their hazard ratio of death at day 14 was 3.10 (p < 0.05) in a multivariate regression model, adjusted for age, sex and comorbidities. In conclusion, CF-carriers are more susceptible to the severe form of COVID-19, showing also higher risk of 14-day death.

SELECTION OF CITATIONS
SEARCH DETAIL