Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 46
Filtrer
1.
Mech Ageing Dev ; 221: 111963, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38986790

RÉSUMÉ

Aging, a complex biological process influenced by genetic, environmental, and pharmacological factors, presents a significant challenge in understanding its underlying mechanisms. In this study, we explored the divergent impacts of metformin treatment on the lifespan and healthspan of young and old C. elegans, demonstrating a intriguing "elixir in youth, poison in elder" phenomenon. By scrutinizing the gene expression changes in response to metformin in young (day 1 of adulthood) and old (days 8) groups, we identified nhr-57 and C46G7.1 as potential modulators of age-specific responses. Notably, nhr-57 and C46G7.1 exhibit contrasting regulation patterns, being up-regulated in young worms but down-regulated in old counterparts following metformin treatment. Functional studies employing knockdown approaches targeting nhr-57, a gene under the control of hif-1 with a documented protective function against pore-forming toxins in C. elegans, and C46G7.1, unveiled their critical roles in modulating lifespan and healthspan, as well as in mediating the biphasic effects of metformin. Furthermore, deletion of hif-1 retarded the influence of metformin, implicating the involvement of hif-1/nhr-57 in age-specific drug responses. These findings underscored the necessity of deciphering the mechanisms governing age-related susceptibility to pharmacological agents to tailor interventions for promoting successful aging.


Sujet(s)
Protéines de Caenorhabditis elegans , Caenorhabditis elegans , Longévité , Metformine , Animaux , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Caenorhabditis elegans/métabolisme , Metformine/pharmacologie , Longévité/effets des médicaments et des substances chimiques , Protéines de Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/génétique , Vieillissement/effets des médicaments et des substances chimiques , Hypoglycémiants/pharmacologie
2.
Hypertension ; 81(9): 1895-1909, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38989583

RÉSUMÉ

BACKGROUND: STIM1 (stromal interaction molecule 1) regulates store-operated calcium entry and is involved in pulmonary artery vasoconstriction and pulmonary artery smooth muscle cell proliferation, leading to pulmonary arterial hypertension (PAH). METHODS: Bioinformatics analysis and a 2-stage matched case-control study were conducted to screen for noncoding variants that may potentially affect STIM1 transcriptional regulation in 242 patients with idiopathic PAH and 414 healthy controls. Luciferase reporter assay, real-time quantitative polymerase chain reaction, western blot, 5-ethynyl-2'-deoxyuridine (EdU) assay, and intracellular Ca2+ measurement were performed to study the mechanistic roles of those STIM1 noncoding variants in PAH. RESULTS: Five noncoding variants (rs3794050, rs7934581, rs3750996, rs1561876, and rs3750994) were identified and genotyped using Sanger sequencing. Rs3794050, rs7934581, and rs1561876 were associated with idiopathic PAH (recessive model, all P<0.05). Bioinformatics analysis showed that these 3 noncoding variants possibly affect the enhancer function of STIM1 or the microRNA (miRNA) binding to STIM1. Functional validation performed in HEK293 and pulmonary artery smooth muscle cells demonstrated that the noncoding variant rs1561876-G (STIM1 mutant) had significantly stronger transcriptional activity than the wild-type counterpart, rs1561876-A, by affecting the transcriptional regulatory function of both hsa-miRNA-3140-5p and hsa-miRNA-4766-5p. rs1561876-G enhanced intracellular Ca2+ signaling in human pulmonary artery smooth muscle cells secondary to calcium-sensing receptor activation and promoted proliferation of pulmonary artery smooth muscle cells under both normoxia and hypoxia conditions, suggesting a possible contribution to PAH development. CONCLUSIONS: The potential clinical implications of the 3 noncoding variants of STIM1, rs3794050, rs7934581, and rs1561876, are 2-fold, as they may help predict the risk and prognosis of idiopathic PAH and guide investigations on novel therapeutic pathway(s).


Sujet(s)
Artère pulmonaire , Molécule-1 d'interaction stromale , Humains , Molécule-1 d'interaction stromale/génétique , Molécule-1 d'interaction stromale/métabolisme , Mâle , Femelle , Études cas-témoins , Adulte d'âge moyen , Adulte , Protéines tumorales/génétique , Myocytes du muscle lisse/métabolisme , Hypertension artérielle pulmonaire primitive familiale/génétique , Hypertension artérielle pulmonaire primitive familiale/physiopathologie , Prédisposition génétique à une maladie , Muscles lisses vasculaires/métabolisme , Régulation de l'expression des gènes , Prolifération cellulaire/génétique , Polymorphisme de nucléotide simple
3.
J Appl Toxicol ; 44(10): 1528-1539, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38840409

RÉSUMÉ

Aging and age-related diseases are intricately associated with oxidative stress and inflammation. Nonsteroidal anti-inflammatory drugs (NSAIDs) have shown their promise in mitigating age-related conditions and potentially extending lifespan in various model organisms. However, the efficacy of NSAIDs in older individuals may be influenced by age-related changes in drug metabolism and tolerance, which could result in age-dependent toxicities. This study aimed to evaluate the potential risks of toxicities associated with commonly used NSAIDs (aspirin, ibuprofen, acetaminophen, and indomethacin) on lifespan, healthspan, and oxidative stress levels in both young and old Caenorhabditis elegans. The results revealed that aspirin and ibuprofen were able to extend lifespan in both young and old worms by suppressing ROS generation and enhancing the expression of antioxidant SOD genes. In contrast, acetaminophen and indomeacin accelerated aging process in old worms, leading to oxidative stress damage and reduced resistance to heat stress through the pmk-1/skn-1 pathway. Notably, the harmful effects of acetaminophen and indomeacin were mitigated when pmk-1 was knocked out in the pmk-1(km25) strain. These results underscore the potential lack of benefit from acetaminophen and indomeacin in elderly individuals due to their increased susceptibility to toxicity. Further research is essential to elucidate the underlying mechanisms driving these age-dependent responses and to evaluate the potential risks associated with NSAID use in the elderly population.


Sujet(s)
Vieillissement , Anti-inflammatoires non stéroïdiens , Caenorhabditis elegans , Longévité , Stress oxydatif , Animaux , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Caenorhabditis elegans/génétique , Anti-inflammatoires non stéroïdiens/toxicité , Longévité/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Vieillissement/effets des médicaments et des substances chimiques , Acétaminophène/toxicité , Protéines de Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/métabolisme , Ibuprofène/toxicité , Acide acétylsalicylique/toxicité , Espèces réactives de l'oxygène/métabolisme
4.
Cell Stem Cell ; 31(8): 1187-1202.e8, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38772378

RÉSUMÉ

Cell-based ex vivo gene therapy in solid organs, especially the liver, has proven technically challenging. Here, we report a feasible strategy for the clinical application of hepatocyte therapy. We first generated high-quality autologous hepatocytes through the large-scale expansion of patient-derived hepatocytes. Moreover, the proliferating patient-derived hepatocytes, together with the AAV2.7m8 variant identified through screening, enabled CRISPR-Cas9-mediated targeted integration efficiently, achieving functional correction of pathogenic mutations in FAH or OTC. Importantly, these edited hepatocytes repopulated the injured mouse liver at high repopulation levels and underwent maturation, successfully treating mice with tyrosinemia following transplantation. Our study combines ex vivo large-scale cell expansion and gene editing in patient-derived transplantable hepatocytes, which holds potential for treating human liver diseases.


Sujet(s)
Systèmes CRISPR-Cas , Édition de gène , Thérapie génétique , Hépatocytes , Maladies du foie , Hépatocytes/métabolisme , Hépatocytes/transplantation , Systèmes CRISPR-Cas/génétique , Humains , Animaux , Maladies du foie/thérapie , Maladies du foie/génétique , Maladies du foie/anatomopathologie , Souris , Thérapie génétique/méthodes , Tyrosinémies/thérapie , Tyrosinémies/génétique , Prolifération cellulaire , Hydrolases
5.
Nat Genet ; 56(5): 938-952, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38627596

RÉSUMÉ

Cholestatic liver injuries, characterized by regional damage around the bile ductular region, lack curative therapies and cause considerable mortality. Here we generated a high-definition spatiotemporal atlas of gene expression during cholestatic injury and repair in mice by integrating spatial enhanced resolution omics sequencing and single-cell transcriptomics. Spatiotemporal analyses revealed a key role of cholangiocyte-driven signaling correlating with the periportal damage-repair response. Cholangiocytes express genes related to recruitment and differentiation of lipid-associated macrophages, which generate feedback signals enhancing ductular reaction. Moreover, cholangiocytes express high TGFß in association with the conversion of liver progenitor-like cells into cholangiocytes during injury and the dampened proliferation of periportal hepatocytes during recovery. Notably, Atoh8 restricts hepatocyte proliferation during 3,5-diethoxycarbonyl-1,4-dihydro-collidin damage and is quickly downregulated after injury withdrawal, allowing hepatocytes to respond to growth signals. Our findings lay a keystone for in-depth studies of cellular dynamics and molecular mechanisms of cholestatic injuries, which may further develop into therapies for cholangiopathies.


Sujet(s)
Cholestase , Hépatocytes , Animaux , Souris , Cholestase/génétique , Cholestase/anatomopathologie , Cholestase/métabolisme , Hépatocytes/métabolisme , Foie/métabolisme , Foie/traumatismes , Foie/anatomopathologie , Prolifération cellulaire/génétique , Conduits biliaires/métabolisme , Régénération hépatique/génétique , Souris de lignée C57BL , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Transduction du signal , Mâle , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Transcriptome , Modèles animaux de maladie humaine , Analyse spatio-temporelle
6.
Cell Stem Cell ; 31(4): 484-498.e5, 2024 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-38458193

RÉSUMÉ

Alginate-encapsulated hepatocyte transplantation is a promising strategy to treat liver failure. However, its clinical application was impeded by the lack of primary human hepatocytes and difficulty in controlling their quality. We previously reported proliferating human hepatocytes (ProliHHs). Here, quality-controlled ProliHHs were produced in mass and engineered as liver organoids to improve their maturity. Encapsulated ProliHHs liver organoids (eLO) were intraperitoneally transplanted to treat liver failure animals. Notably, eLO treatment increased the survival of mice with post-hepatectomy liver failure (PHLF) and ameliorated hyperammonemia and hypoglycemia by providing liver functions. Additionally, eLO treatment protected the gut from PHLF-augmented permeability and normalized the increased serum endotoxin and inflammatory response, which facilitated liver regeneration. The therapeutic effect of eLO was additionally proved in acetaminophen-induced liver failure. Furthermore, we performed assessments of toxicity and biodistribution, demonstrating that eLO had no adverse effects on animals and remained non-tumorigenic.


Sujet(s)
Défaillance hépatique aigüe , Défaillance hépatique , Humains , Souris , Animaux , Défaillance hépatique aigüe/thérapie , Défaillance hépatique aigüe/induit chimiquement , Distribution tissulaire , Cellules cultivées , Hépatocytes , Foie , Défaillance hépatique/thérapie , Défaillance hépatique/métabolisme , Organoïdes/métabolisme
7.
Am J Transplant ; 23(12): 1832-1844, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37532180

RÉSUMÉ

Hepatocyte transplantation has the potential to treat acute liver failure and correct liver-based metabolic disorders. Proliferating human hepatocytes (ProliHHs) provide a large-scale source as an alternative to primary human hepatocytes. However, host rejection led to inefficient graft survival and function, which hindered the clinical application of cell therapy. Herein, we employed the lentiviral system to overexpress immunomodulatory factors programmed death-ligand 1 (cluster of differentiation 274) (CD274) and cluster of differentiation 47 (CD47) in ProliHHs. CD47+274 overexpression inhibited macrophage and T cell responses in vitro. After transplantation into mice via the spleen without immunosuppression, CD47+274 ProliHHs accumulation in the liver significantly increased for 48 hours compared with ProliHHs. Consistent with the in vitro results, CD47+274 ProliHHs were less aggregated and infiltrated by macrophages and also recruited fewer T cells in the liver. Seven days after transplantation, the human albumin level of engineered ProliHHs doubled compared with control group. CD47+274 ProliHHs further ameliorated the liver injury induced using concanavalin A. Overall, our results suggested CD47+274 overexpression reduced innate and adaptive immune responses during hepatocyte transplantation, and the survival rate and graft function of transplanted hepatocyte-like cells were all significantly improved.


Sujet(s)
Antigènes CD47 , Maladies du foie , Animaux , Humains , Souris , Antigène CD274/métabolisme , Hépatocytes , Immunité , Maladies du foie/métabolisme
8.
Cell Stem Cell ; 30(5): 617-631.e8, 2023 05 04.
Article de Anglais | MEDLINE | ID: mdl-37059100

RÉSUMÉ

Liver resection is the first-line treatment for primary liver cancers, providing the potential for a cure. However, concerns about post-hepatectomy liver failure (PHLF), a leading cause of death following extended liver resection, have restricted the population of eligible patients. Here, we engineered a clinical-grade bioartificial liver (BAL) device employing human-induced hepatocytes (hiHeps) manufactured under GMP conditions. In a porcine PHLF model, the hiHep-BAL treatment showed a remarkable survival benefit. On top of the supportive function, hiHep-BAL treatment restored functions, specifically ammonia detoxification, of the remnant liver and facilitated liver regeneration. Notably, an investigator-initiated study in seven patients with extended liver resection demonstrated that hiHep-BAL treatment was well tolerated and associated with improved liver function and liver regeneration, meeting the primary outcome of safety and feasibility. These encouraging results warrant further testing of hiHep-BAL for PHLF, the success of which would broaden the population of patients eligible for liver resection.


Sujet(s)
Défaillance hépatique , Foie artificiel , Humains , Animaux , Suidae , Hépatocytes , Défaillance hépatique/chirurgie , Régénération hépatique
9.
Cell Stem Cell ; 30(3): 283-299.e9, 2023 03 02.
Article de Anglais | MEDLINE | ID: mdl-36787740

RÉSUMÉ

Stem cell-independent reprogramming of differentiated cells has recently been identified as an important paradigm for repairing injured tissues. Following periportal injury, mature hepatocytes re-activate reprogramming/progenitor-related genes (RRGs) and dedifferentiate into liver progenitor-like cells (LPLCs) in both mice and humans, which contribute remarkably to regeneration. However, it remains unknown which and how external factors trigger hepatocyte reprogramming. Here, by employing single-cell transcriptional profiling and lineage-specific deletion tools, we uncovered that periportal-specific LPLC formation was initiated by regionally activated Kupffer cells but not peripheral monocyte-derived macrophages. Unexpectedly, using in vivo screening, the proinflammatory factor IL-6 was identified as the niche signal repurposed for RRG induction via STAT3 activation, which drove RRG expression through binding to their pre-accessible enhancers. Notably, RRGs were activated through injury-specific rather than liver embryogenesis-related enhancers. Collectively, these findings depict an injury-specific niche signal and the inflammation-mediated transcription in driving the conversion of hepatocytes into a progenitor phenotype.


Sujet(s)
Interleukine-6 , Cellules de Küpffer , Animaux , Humains , Souris , Différenciation cellulaire , Hépatocytes/métabolisme , Interleukine-6/métabolisme , Cellules de Küpffer/physiologie , Foie , Régénération hépatique/physiologie
10.
Hepatology ; 76(6): 1690-1705, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-35229337

RÉSUMÉ

BACKGROUND AND AIMS: Hepatocyte transplantation has been demonstrated to be effective to treat liver metabolic disease and acute liver failure. Nevertheless, the shortage of donor hepatocytes restrained its application in clinics. To expand human hepatocytes at a large scale, several dedifferentiation-based protocols have been established, including proliferating human hepatocytes (ProliHH). However, the decreased transplantation efficiency of these cells after long-term expansion largely impedes their application. APPROACH AND RESULTS: We found that accompanied with dedifferentiation, long-term cultured ProliHH (lc-ProliHH) up-regulated a panel of chemokines and cytokines related to innate immunity, which were referred to as dedifferentiation-associated inflammatory factors (DAIF). DAIF elicited excessive macrophage responses, accounting for the elimination of lc-ProliHH specifically during engraftment. Two possible strategies to increase ProliHH transplantation were then characterized. Blockage of innate immune response by dexamethasone reverted the engraftment and repopulation of lc-ProliHH to a level comparable to primary hepatocytes, resulting in improved liver function and a better survival of fumarylacetoacetate hydrolase-deficient mice. Alternatively, rematuration of lc-ProliHH as organoids reduced the expression of DAIF and led to markedly improved engraftment. CONCLUSIONS: These results revealed that lc-ProliHH triggers exacerbated macrophage activation by DAIF and provided potential solutions for clinical transplantation of lc-ProliHH.


Sujet(s)
Hépatocytes , Foie , Humains , Souris , Animaux , Hépatocytes/métabolisme , Foie/métabolisme , Cytokines/métabolisme , Chimiokines/métabolisme , Macrophages/métabolisme
11.
EClinicalMedicine ; 43: 101200, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-35128361

RÉSUMÉ

BACKGROUND: Intrauterine adhesions (IUA) develop in up to 20% of women with a history of abortion. After hysteroscopic adhesiolysis, balloon stents are usually placed for seven days to prevent recurrence. The efficacy of prolonged use (30 days) of balloon stents has not been determined. METHODS: The trial was conducted from June 2019 to March 2021. Ninety-one patients who underwent hysteroscopic adhesiolysis for moderate or severe IUA were randomized to receive a 30-day placement of a balloon stent (n = 44) or an intrauterine device (IUD) (n = 47). The primary outcomes were the ongoing pregnancy and miscarriage rates assessed at 15-19 months. The secondary outcomes were the recurrence of IUA and the American Fertility Society (AFS) intrauterine adhesion scores at the first and second hysteroscopies, the diagnosis of new chronic endometritis at the second-look hysteroscopy, and the vaginal/uterine microbiome assessed using 16S rRNA sequencing. The trial was registered at Chinese Clinical Trial Registry (ChiCTR1900023306). FINDINGS: The ongoing pregnancy rates (balloon 56·4% versus IUD 57·1%) and miscarriage rates (balloon 10·3% versus IUD 22·9%) were not significantly different between the groups. No differences in the recurrence of IUA, reduction of AFS scores, or new endometritis rates were detected. The bacterial load in the uterus and vagina increased in the IUD group but not in the balloon group. INTERPRETATION: Balloon placement has a similar effect on ongoing pregnancy rates as intrauterine device (IUD) placement. Patients who underwent balloon placement had a lower miscarriage rate, although the difference was not significant. There were no differences in the recurrence rate of IUA, reduction of American Fertility Society scores, or rate of new chronic endometritis. The balloon stent has less effects on the uterine microbiota. FUNDING: National Natural Science Foundation of China (81802593).

12.
Int J Nanomedicine ; 16: 6719-6747, 2021.
Article de Anglais | MEDLINE | ID: mdl-34621124

RÉSUMÉ

Despite several recent advances, current therapy and prevention strategies for myocardial infarction are far from satisfactory, owing to limitations in their applicability and treatment effects. Nanoparticles (NPs) enable the targeted and stable delivery of therapeutic compounds, enhance tissue engineering processes, and regulate the behaviour of transplants such as stem cells. Thus, NPs may be more effective than other mechanisms, and may minimize potential adverse effects. This review provides evidence for the view that function-oriented systems are more practical than traditional material-based systems; it also summarizes the latest advances in NP-based strategies for the treatment and prevention of myocardial infarction.


Sujet(s)
Infarctus du myocarde , Nanoparticules , Systèmes de délivrance de médicaments , Humains , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/prévention et contrôle , Nanoparticules/usage thérapeutique
14.
Acta Pharmacol Sin ; 42(12): 2082-2093, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-33654219

RÉSUMÉ

Sulfur mustard (SM) is a highly toxic chemical warfare agent that causes acute lung injury (ALI) and/or acute respiratory distress syndrome (ARDS). There are no effective therapeutic treatments or antidotes available currently to counteract its toxic effects. Our previous study shows that bone marrow-derived mesenchymal stromal cells (BMSCs) could exert therapeutic effects against SM-induced lung injury. In this study, we explored the therapeutic potential of BMSC-derived exosomes (BMSC-Exs) against ALI and the underlying mechanisms. ALI was induced in mice by injection of SM (30 mg/kg, sc) at their medial and dorsal surfaces. BMSC-Exs (20 µg/kg in 200 µL PBS, iv) were injected for a 5-day period after SM exposure. We showed that BMSC-Exs administration caused a protective effect against pulmonary edema. Using a lung epithelial cell barrier model, BMSC-Exs (10, 20, 40 µg) dose-dependently inhibited SM-induced cell apoptosis and promoted the recovery of epithelial barrier function by facilitating the expression and relocalization of junction proteins (E-cadherin, claudin-1, occludin, and ZO-1). We further demonstrated that BMSC-Exs protected against apoptosis and promoted the restoration of barrier function against SM through upregulating G protein-coupled receptor family C group 5 type A (GPRC5A), a retinoic acid target gene predominately expressed in the epithelial cells of the lung. Knockdown of GPRC5A reduced the antiapoptotic and barrier regeneration abilities of BMSC-Exs and diminished their therapeutic effects in vitro and in vivo. BMSC-Exs-caused upregulation of GPRC5A promoted the expression of Bcl-2 and junction proteins via regulating the YAP pathway. In summary, BMSC-Exs treatment exerts protective effects against SM-induced ALI by promoting alveolar epithelial barrier repair and may be an alternative approach to stem cell-based therapy.


Sujet(s)
Lésion pulmonaire aigüe/thérapie , Exosomes/transplantation , Cellules souches mésenchymateuses/cytologie , Transduction du signal/effets des médicaments et des substances chimiques , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/métabolisme , Animaux , Apoptose/physiologie , Lignée cellulaire , Cellules épithéliales/métabolisme , Techniques de knock-out de gènes , Poumon/métabolisme , Poumon/anatomopathologie , Mâle , Souris de lignée ICR , Souris knockout , Gaz moutarde , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Protéines de signalisation YAP/métabolisme
15.
Dis Markers ; 2020: 3178642, 2020.
Article de Anglais | MEDLINE | ID: mdl-32670434

RÉSUMÉ

BACKGROUND: Exosomes exist in almost all body fluid and contain diverse biological contents which may be reflective of disease state. Circular RNAs (circRNAs) are stable in structure and have a long half-life in exosomes without degradation, thus making them reliable biomarkers. However, the potential of exosomal circRNAs as biomarkers of coronary artery disease (CAD) remains to be established. Here, we aimed to investigate the expression levels and the potential use of exosomal circRNAs as diagnostic biomarkers for CAD. METHODS: CircRNA expression levels in exosomes obtained from three plasma samples of CAD patients and three paired controls were analyzed using RNA sequencing. Exosomal circRNAs obtained in the profiling phase were then verified in two-center validation cohorts. Finally, the ability of exosomal circRNAs, adjusting for Framingham Heart Study (FHS) risk factors, was determined to discriminate between CAD patients and non-CAD controls. RESULTS: 355 circRNAs were differentially expressed between these two groups: 164 were upregulated, and 191 were downregulated. Here, we selected the potential circRNAs (fold change > 4, P < 0.05) as candidate biomarkers for further validation. Our data showed that only hsa_circ_0005540 was significantly associated with CAD (P < 0.0001). After adjustment for risk factors, hsa_circ_0005540 showed a high discriminatory power for CAD in ROC analyses (AUC = 0.853; 95%confidence interval (CI) = 0.799 - 0.906, P < 0.001). CONCLUSION: Our results suggest that plasma exosomal hsa_circ_0005540 can be used as a promising diagnostic biomarker of CAD.


Sujet(s)
Marqueurs biologiques/sang , Maladie des artères coronaires/diagnostic , Exosomes/génétique , Analyse de profil d'expression de gènes/méthodes , ARN circulaire/sang , Adulte , Sujet âgé , Études cas-témoins , Maladie des artères coronaires/sang , Diagnostic précoce , Femelle , Séquençage nucléotidique à haut débit , Humains , Mâle , Adulte d'âge moyen , Analyse de séquence d'ARN , Régulation positive
16.
Aging (Albany NY) ; 12(13): 13076-13089, 2020 07 02.
Article de Anglais | MEDLINE | ID: mdl-32614786

RÉSUMÉ

CircFOXO3 plays an important role in the pathogenesis of coronary artery disease (CAD). Single nucleotide polymorphisms (SNPs) at circRNA flanking introns may change its back-splicing and influence circRNA formation. Here, we aimed to investigate the influence of the polymorphisms at the circFOXO3 flanking introns on individual susceptibility to CAD. A total of 1185 individuals were included in the case-control study. In a multivariate logistic regression analysis, we determined that the rs12196996 G variant was significantly associated with increased CAD risk (OR = 1.36, P = 0.014). A similar trend of the association was observed in the recessive model (OR = 2.57, P = 0.003). Stratified analysis revealed a more significant association with CAD risk among younger subjects and non-smokers. Consistent with these results, the haplotype rs12196996G-rs9398171C containing rs12196996G allele was also associated with increased CAD risk (OR = 1.31, P = 0.013). Further investigation revealed that the rs12196996 GG genotype was associated with decreased circFOXO3 expression, but not linear FOXO3 levels. Taken together, our data provide the first evidence that the rs12196996 polymorphism at the circFOXO3 gene flanking intron is associated with CAD risk in the Chinese Han population, which is probably due to influence circFOXO3 levels.


Sujet(s)
Maladie des artères coronaires , Protéine O3 à motif en tête de fourche/génétique , Prédisposition génétique à une maladie , Polymorphisme de nucléotide simple/génétique , ARN circulaire/génétique , Sujet âgé , Asiatiques/génétique , Études cas-témoins , Chine , Maladie des artères coronaires/épidémiologie , Maladie des artères coronaires/génétique , Femelle , Prédisposition génétique à une maladie/épidémiologie , Prédisposition génétique à une maladie/génétique , Humains , Introns/génétique , Mâle , Adulte d'âge moyen , Facteurs de risque
17.
Hum Gene Ther ; 31(5-6): 286-296, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-32013585

RÉSUMÉ

Osteonecrosis of the femoral head (ONFH) is a common and disabling joint disease. Although there is no clear consensus on the complex pathogenic mechanism of ONFH, trauma, abuse of glucocorticoids, and alcoholism are implicated in its etiology. The therapeutic strategies are still limited, and the clinical outcomes are not satisfactory. Mesenchymal stem cells (MSCs) have been shown to exert a positive impact on ONFH in preclinical experiments and clinical trials. The beneficial properties of MSCs are due, at least in part, to their ability to home to the injured tissue, secretion of paracrine signaling molecules, and multipotentiality. Nevertheless, the regenerative capacity of transplanted cells is impaired by the hostile environment of necrotic tissue in vivo, limiting their clinical efficacy. Recently, genetic engineering has been introduced as an attractive strategy to improve the regenerative properties of MSCs in the treatment of early-stage ONFH. This review summarizes the function of several genes used in the engineering of MSCs for the treatment of ONFH. Further, current challenges and future perspectives of genetic manipulation of MSCs are discussed. The notion of genetically engineered MSCs functioning as a "factory" that can produce a significant amount of multipotent and patient-specific therapeutic product is emphasized.


Sujet(s)
Nécrose de la tête fémorale/génétique , Nécrose de la tête fémorale/thérapie , Thérapie génétique/méthodes , Cellules souches mésenchymateuses/physiologie , Animaux , Chimiokines/physiologie , Facteur de croissance fibroblastique de type 2/physiologie , Génie génétique , Facteur de croissance des hépatocytes/physiologie , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/physiologie , Protéines et peptides de signalisation intercellulaire/physiologie , Ostéogenèse , Facteur de croissance endothéliale vasculaire de type A/physiologie
18.
World J Pediatr ; 16(3): 305-313, 2020 Jun.
Article de Anglais | MEDLINE | ID: mdl-31912317

RÉSUMÉ

BACKGROUND: Hearing impairment is one of the most common birth defects in children. Universal newborn hearing screenings have been performed for 19 years in Guangdong province, China. A screening/diagnosis/intervention system has gradually been put in place. Over the past 10 years, a relatively complete data management system had been established. In the present study, an etiological analysis of newborn cases that failed the initial and follow-up screenings was performed. METHODS: The nature and degree of hearing impairment in newborns were confirmed by a set of procedures performed at the time of initial hearing screening, rescreening and final hearing diagnosis. Then, multiple examinations were performed to explore the associated etiology. RESULTS: Over a period of 10 years, 720 children were diagnosed with newborn hearing loss. Among these children, 445 (61.81%) children had a clearly identified cause, which included genetic factor(s) (30.56%), secretory otitis media (13.30%), maternal rubella virus infection during pregnancy (5.83%), inner ear malformations (4.86%), maternal human cytomegalovirus infection during pregnancy (2.92%), malformation of the middle ear ossicular chain (2.50%) and auditory neuropathy (1.81%). In addition, 275 cases of sensorineural hearing loss of unknown etiology accounted for 38.19% of the children surveyed. CONCLUSIONS: Long-term follow-up is needed to detect delayed hearing impairment and auditory development in children. The need for long-term follow-up should be taken into account when designing an intervention strategy. Furthermore, the use of the deafness gene chip should further elucidate the etiology of neonatal hearing impairment.


Sujet(s)
Perte d'audition/congénital , Perte d'audition/étiologie , Dépistage néonatal , Chine/épidémiologie , Évolution de la maladie , Femelle , Études de suivi , Perte d'audition/épidémiologie , Humains , Nouveau-né , Mâle , Études rétrospectives
19.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-828024

RÉSUMÉ

To scientifically evaluate the intervention effect of Chinese medicine preventive administration(combined use of Huo-xiang Zhengqi Oral Liquid and Jinhao Jiere Granules) on community population in the case of coronavirus disease 2019(COVID-19), a large cohort, prospective, randomized, and parallel-controlled clinical study was conducted. Total 22 065 subjects were included and randomly divided into 2 groups. The non-intervention group was given health guidance only, while the traditional Chinese medicine(TCM) intervention group was given two coordinated TCM in addition to health guidance. The medical instructions were as follows. Huoxiang Zhengqi Oral Liquid: oral before meals, 10 mL/time, 2 times/day, a course of 5 days. Jinhao Jiere Granules: dissolve in boiling water and take after meals, 8 g/time, 2 times/day, a course of 5 days, followed up for 14 days, respectively. The study found that with the intake of medication, the incidence rate of TCM intervention group was basically maintained at a low and continuous stable level(0.01%-0.02%), while the non-intervention group showed an overall trend of continuous growth(0.02%-0.18%) from 3 to 14 days. No suspected or confirmed COVID-19 case occurred in either group. There were 2 cases of colds in the TCM intervention group and 26 cases in the non-intervention group. The incidence of colds in the TCM intervention group was significantly lower(P<0.05) than that in the non-intervention group. In the population of 16-60 years old, the incidence rate of non-intervention and intervention groups were 0.01% and 0.25%, respectively. The difference of colds incidence between the two groups was statistically significant(P<0.05). In the population older than 60 years old, they were 0.04% and 0.21%, respectively. The incidence of colds in the non-intervention group was higher than that in the intervention group, but not reaching statistical difference. The protection rate of TCM for the whole population was 91.8%, especially for the population of age 16-60(95.0%). It was suggested that TCM intervention(combined use of Huoxiang Zhengqi Oral Liquid and Jinhao Jiere Granules) could effectively protect community residents against respiratory diseases, such as colds, which was worthy of promotion in the community. In addition, in terms of safety, the incidence of adverse events and adverse reactions in the TCM intervention group was relatively low, which was basically consistent with the drug instructions.


Sujet(s)
Adolescent , Adulte , Humains , Adulte d'âge moyen , Jeune adulte , Betacoronavirus , Infections à coronavirus , Traitement médicamenteux , Médicaments issus de plantes chinoises , Médecine traditionnelle chinoise , Pandémies , Pneumopathie virale , Traitement médicamenteux , Études prospectives
20.
Cancer Cell ; 36(2): 179-193.e11, 2019 08 12.
Article de Anglais | MEDLINE | ID: mdl-31378681

RÉSUMÉ

Liver cancers are highly heterogeneous with poor prognosis and drug response. A better understanding between genetic alterations and drug responses would facilitate precision treatment for liver cancers. To characterize the landscape of pharmacogenomic interactions in liver cancers, we developed a protocol to establish human liver cancer cell models at a success rate of around 50% and generated the Liver Cancer Model Repository (LIMORE) with 81 cell models. LIMORE represented genomic and transcriptomic heterogeneity of primary cancers. Interrogation of the pharmacogenomic landscape of LIMORE discovered unexplored gene-drug associations, including synthetic lethalities to prevalent alterations in liver cancers. Moreover, predictive biomarker candidates were suggested for the selection of sorafenib-responding patients. LIMORE provides a rich resource facilitating drug discovery in liver cancers.


Sujet(s)
Antinéoplasiques/pharmacologie , Marqueurs biologiques tumoraux/génétique , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Variants pharmacogénomiques , Inhibiteurs de protéines kinases/pharmacologie , Sorafénib/pharmacologie , Animaux , Asiatiques/génétique , Carcinome hépatocellulaire/ethnologie , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale , Prise de décision clinique , Bases de données génétiques , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Hétérogénéité génétique , Prédisposition génétique à une maladie , Séquençage nucléotidique à haut débit , Humains , Tumeurs du foie/ethnologie , Tumeurs du foie/anatomopathologie , Mâle , Souris de lignée BALB C , Souris de lignée NOD , Souris nude , Souris SCID , Sélection de patients , Test pharmacogénomique , Phénotype , Médecine de précision , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE