Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 68
Filtrer
1.
Environ Pollut ; 360: 124536, 2024 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-39029862

RÉSUMÉ

Tri (2-Ethylhexyl) phosphate (TEHP), widely used as a fire retardant and plasticizer, has been commonly found in the environment. Its potential health-related risks, especially reproductive toxicity, have aroused concern. However, the potential cellular mechanisms remain unexplored. In this study, we aimed to investigate the molecular mechanisms underlying TEHP-caused cell damage in Sertoli cells, which play a crucial role in supporting spermatogenesis. Our findings indicate that TEHP induces apoptosis in 15P-1 mouse Sertoli cells. Subsequently, we conducted RNA sequencing analyses, which suggested that ER stress, autophagy, and MAPK-related pathways may participate in TEHP-induced cytotoxicity. Furthermore, we demonstrated that TEHP triggers ER stress, activates p38 MAPK, and inhibits autophagy flux. Then, we showed that the inhibition of ER stress or p38 MAPK activation attenuates TEHP-induced apoptosis, while the inhibition of autophagy flux is responsible for TEHP-induced apoptosis. These results collectively reveal that TEHP induces ER stress, activates p38, and inhibits autophagy flux, ultimately leading to apoptosis in Sertoli cells. These shed light on the molecular mechanisms underlying TEHP-associated testicular toxicity.


Sujet(s)
Apoptose , Autophagie , Stress du réticulum endoplasmique , Cellules de Sertoli , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Cellules de Sertoli/effets des médicaments et des substances chimiques , Cellules de Sertoli/métabolisme , Animaux , Mâle , Autophagie/effets des médicaments et des substances chimiques , Souris , Apoptose/effets des médicaments et des substances chimiques , Analyse de séquence d'ARN , p38 Mitogen-Activated Protein Kinases/métabolisme , p38 Mitogen-Activated Protein Kinases/génétique , Ignifuges/toxicité , Plastifiants/toxicité , Phtalate de bis[2-éthylhexyle]/toxicité , Phtalate de bis[2-éthylhexyle]/analogues et dérivés
2.
J Pediatr Endocrinol Metab ; 37(8): 734-740, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39026465

RÉSUMÉ

OBJECTIVES: Pseudohypoparathyroidism (PHP) comprises a cluster of heterogeneous diseases characterized by hypocalcemia and hyperphosphatemia due to parathyroid hormone (PTH) resistance. PHP type 1B (PHP1B) is caused by heterozygous maternal deletions within GNAS or STX16. STX16 exon 2-6 deletion is commonly observed in autosomal dominant (AD)-PHP1B, while sporadic PHP1B commonly results from methylation abnormalities of maternal differentially methylated regions and remains unclear at the molecular level. CASE PRESENTATION: A 39-year-old male patient with PHP1B, who had his first seizure at 15 years of age, presented to our hospital. The methylation-specific multiplex ligation-dependent probe amplification results showed a half-reduced copy number of STX16 exon 5-7 and loss of methylation at GNAS exon A/B. His mother also had a half-reduced copy number of STX16 exon 5-7 but with normal methylation of GNAS. His father has a normal copy number of STX16 and normal methylation of GNAS. CONCLUSIONS: For the recognition and early diagnosis of this kind of disease, here we report the clinical symptoms, auxiliary examinations, genetic testing characteristics, and treatment of the patient.


Sujet(s)
Exons , Pseudohypoparathyroïdie , Syntaxine-16 , Humains , Mâle , Pseudohypoparathyroïdie/génétique , Pseudohypoparathyroïdie/complications , Adulte , Syntaxine-16/génétique , Exons/génétique , Délétion de séquence , Sous-unités alpha Gs des protéines G/génétique , Pronostic , Chromogranine/génétique
3.
Adv Biol (Weinh) ; : e2400120, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38864263

RÉSUMÉ

Triptolide (TP), an active component isolated from the traditional Chinese herb Tripterygium wilfordii Hook F (TWHF), shows great promise for treating inflammation-related diseases. However, its potential nephrotoxic effects remain concerning. The mechanism underlying TP-induced nephrotoxicity is inadequately elucidated, particularly at single-cell resolution. Hence, single-cell RNA sequencing (scRNA-seq) of kidney tissues from control and TP-treated mice is performed to generate a thorough description of the renal cell atlas upon TP treatment. Heterogeneous responses of nephron epithelial cells are observed after TP exposure, attributing differential susceptibility of cell subtypes to excessive reactive oxygen species and increased inflammatory responses. Moreover, TP disrupts vascular function by activating endothelial cell immunity and damaging fibroblasts. Severe immune cell damage and the activation of pro-inflammatory Macro_C1 cells are also observed with TP treatment. Additionally, ligand-receptor crosstalk analysis reveals that the SPP1 (osteopontin) signaling pathway targeting Macro_C1 cells is triggered by TP treatment, which may promote the infiltration of Macro_C1 cells to exacerbate renal toxicity. Overall, this study provides comprehensive information on the transcriptomic profiles and cellular composition of TP-associated nephrotoxicity at single-cell resolution, which can strengthen the understanding of the pathogenesis of TP-induced nephrotoxicity and provide valuable clues for the discovery of new therapeutic targets to ameliorate TP-associated nephrotoxicity.

4.
J Environ Sci (China) ; 145: 164-179, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-38844317

RÉSUMÉ

The occurrence of poisoning incidents caused by cyanobacterial blooms has aroused wide public concern. Microcystin-leucine arginine (MC-LR) is a well-established toxin produced by cyanobacterial blooms, which is widely distributed in eutrophic waters. MC-LR is not only hazardous to the water environment but also exerts multiple toxic effects including liver toxicity in both humans and animals. However, the underlying mechanisms of MC-LR-induced liver toxicity are unclear. Herein, we used advanced single-cell RNA sequencing technology to characterize MC-LR-induced liver injury in mice. We established the first single-cell atlas of mouse livers in response to MC-LR. Our results showed that the differentially expressed genes and pathways in diverse cell types of liver tissues of mice treated with MC-LR are highly heterogeneous. Deep analysis showed that MC-LR induced an increase in a subpopulation of hepatocytes that highly express Gstm3, which potentially contributed to hepatocyte apoptosis in response to MC-LR. Moreover, MC-LR increased the proportion and multiple subtypes of Kupffer cells with M1 phenotypes and highly expressed proinflammatory genes. Furthermore, the MC-LR increased several subtypes of CD8+ T cells with highly expressed multiple cytokines and chemokines. Overall, apart from directly inducing hepatocytes apoptosis, MC-LR activated proinflammatory Kupffer cell and CD8+ T cells, and their interaction may constitute a hostile microenvironment that contributes to liver injury. Our findings not only present novel insight into underlying molecular mechanisms but also provide a valuable resource and foundation for additional discovery of MC-LR-induced liver toxicity.


Sujet(s)
Microcystines , Analyse de séquence d'ARN , Microcystines/toxicité , Animaux , Souris , Foie/effets des médicaments et des substances chimiques , Toxines de la flore et de la faune marines/toxicité , Leucine , Hépatocytes/effets des médicaments et des substances chimiques , Lésions hépatiques dues aux substances
5.
Ecotoxicol Environ Saf ; 279: 116462, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38776784

RÉSUMÉ

Tris (2-ethylhexyl) phosphate (TEHP) is a frequently used organophosphorus flame retardant with significant ecotoxicity and widespread human exposure. Recent research indicates that TEHP has reproductive toxicity. However, the precise cell mechanism is not enough understood. Here, by using testicular mesenchymal stromal TM3 cells as a model, we reveal that TEHP induces apoptosis. Then RNA sequencing analysis, immunofluorescence, and western blotting results show that THEP inhibits autophagy flux and enhances endoplasmic reticulum (ER) stress. Moreover, the activation of the ER stress is critical for TEHP-induced cell injury. Interestingly, TEHP-induced ER stress is contributed to autophagic flux inhibition. Furthermore, pharmacological inhibition of autophagy aggravates, and activation of autophagy attenuates TEHP-induced apoptosis. In summary, these findings indicate that TEHP triggers apoptosis in mouse TM3 cells through ER stress activation and autophagy flux inhibition, offering a new perspective on the mechanisms underlying TEHP-induced interstitial cytotoxicity in the mouse testis.


Sujet(s)
Apoptose , Autophagie , Stress du réticulum endoplasmique , Ignifuges , Cellules de Leydig , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Animaux , Mâle , Cellules de Leydig/effets des médicaments et des substances chimiques , Souris , Apoptose/effets des médicaments et des substances chimiques , Ignifuges/toxicité , Lignée cellulaire
6.
Sci Total Environ ; 930: 172582, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38649052

RÉSUMÉ

Perfluoroalkyl sulfonate (PFOS) is a commonly used chemical compound that often found in materials such as waterproofing agents, food packaging, and fire retardants. Known for its stability and persistence in the environment, PFOS can enter the human body through various pathways, including water and the food chain, raising concerns about its potential harm to human health. Previous studies have suggested a cardiac toxicity of PFOS, but the specific cellular mechanisms remained unclear. Here, by using AC16 cardiomyocyte as a model to investigate the molecular mechanisms potential the cardiac toxicity of PFOS. Our findings revealed that PFOS exposure reduced cell viability and induces apoptosis in human cardiomyocyte. Proteomic analysis and molecular biological techniques showed that the Endoplasmic Reticulum (ER) stress-related pathways were activated, while the cellular autophagy flux was inhibited in PFOS-exposed cells. Subsequently, we employed strategies such as autophagy activation and ER stress inhibition to alleviate the PFOS-induced apoptosis in AC16 cells. These results collectively suggest that PFOS-induced ER stress activation and autophagy flux inhibition contribute to cardiomyocyte apoptosis, providing new insights into the mechanisms of PFOS-induced cardiomyocyte toxicity.


Sujet(s)
Acides alcanesulfoniques , Apoptose , Autophagie , Stress du réticulum endoplasmique , Fluorocarbones , Myocytes cardiaques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Fluorocarbones/toxicité , Acides alcanesulfoniques/toxicité , Humains , Lignée cellulaire , Polluants environnementaux/toxicité
7.
Environ Int ; 185: 108543, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38452464

RÉSUMÉ

Exposure to environmentally hazardous substances is recognized as a significant risk factor for neurological associated disorders. Among these substances, polystyrene microplastics (PS-MPs), widely utilized in various consumer products, have been reported to exhibit neurotoxicity. However, the potential association of PS-MPs with abnormal anxiety behaviors, along with the underlying molecular mechanisms and key proteins involved, remains insufficiently explored. Here, we delineated the potential mechanisms of PS-MPs-induced anxiety through proteomics and molecular investigations. We characterized the PS-MPs, observed their accumulation in the brain, leading to anxiety-like behavior in mice, which is correlated with microglia activation and pro-inflammatory response. Consistent with these findings, our studies on BV2 microglia cells showed that PS-MPs activated NF-κB-mediated inflammation resulting in the upregulation of pro-inflammatory cytokines such as TNFα and IL-1ß. Of particular significance, HRAS was identified as a key factor in the PS-MPs induced pro-inflammatory response through whole proteomics analysis, and knockdown of H-ras effectively inhibited PS-MPs induced PERK-NF-κB activation and associated pro-inflammatory response in microglia cells. Collectively, our findings highlight that PS-MPs induce anxiety of mice via the activation of the HRAS-derived PERK-NF-κB pathway in microlglia. Our results contribute valuable insights into the molecular mechanisms of PS-MPs-induced anxiety, and may offer implications for addressing neurotoxicity and prevention the adverse effects of environmentally hazardous substances, including microplastics.


Sujet(s)
Facteur de transcription NF-kappa B , Syndromes neurotoxiques , Animaux , Souris , Anxiété/induit chimiquement , Produits dangereux , Microplastiques/toxicité , Matières plastiques , Polystyrènes/toxicité
8.
Cancer Lett ; 587: 216622, 2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38246224

RÉSUMÉ

Triptolide, a natural bioactive compound derived from herbal medicine Tripterygium wilfordii, has multiple biological activities including anti-cancer effect, which is being tested in clinical trials for treating cancers. However, the exact mechanism by which Triptolide exerts its cytotoxic effects, particularly its specific protein targets, remains unclear. Here, we show that Triptolide effectively induces cytotoxicity in gastric cancer cells by increasing reactive oxygen species (ROS) levels. Further investigations reveal that ROS accumulation contributes to the induction of Endoplasmic Reticulum (ER) stress, and subsequently autophagy induction in response to Triptolide. Meanwhile, this autophagy is cytoprotective. Interestingly, through activity-based protein profiling (ABPP) approach, we identify peroxiredoxins-2 (PRDX2), a component of the key enzyme systems that act in the defense against oxidative stress and protect cells against hydroperoxides, as direct binding target of Triptolide. By covalently binding to PRDX2 to inhibit its antioxidant activity, Triptolide increases ROS levels. Moreover, overexpression of PRDX2 inhibits and knockdown of the expression of PRDX2 increases Triptolide-induced apoptosis. Collectively, these results indicate PRDX2 as a direct target of Triptolides for inducing apoptosis. Our results not only provide novel insight into the underlying mechanisms of Triptolide-induced cytotoxic effects, but also indicate PRDX2 as a promising potential therapeutic target for developing anti-gastric cancer agents.


Sujet(s)
Diterpènes , Phénanthrènes , Tumeurs de l'estomac , Humains , Espèces réactives de l'oxygène/métabolisme , Tumeurs de l'estomac/traitement médicamenteux , Peroxirédoxines/génétique , Diterpènes/pharmacologie , Phénanthrènes/pharmacologie , Autophagie , Apoptose , Composés époxy/pharmacologie
9.
Sci Total Environ ; 917: 169861, 2024 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-38185161

RÉSUMÉ

Perfluorooctanoic acid (PFOA) is a man-made chemical broadly distributed in various ecological environment and human bodies, which poses potential health risks. Its toxicity, especially the male reproduction toxicity has drawn increasing attention due to declining birth rates in recent years. However, how PFOA induces male reproductive toxicity remains unclear. Here, we characterize PFOA-induced cell injury and reveal the underlying mechanism in mouse Leydig cells, which are critical to spermatogenesis in the testes. We show that PFOA induces cell injury as evidenced by reduced cell viability, cell morphology changes and apoptosis induction. RNA-sequencing analysis reveals that PFOA-induced cell injury is correlated with compromised autophagy and activated endoplasmic reticulum (ER) stress, two conserved biological processes required for regulating cellular homeostasis. Mechanistic analysis shows that PFOA inhibits autophagosomes formation, and activation of autophagy rescues PFOA-induced apoptosis. Additionally, PFOA activates ER stress, and pharmacological inhibition of ER stress attenuates PFOA-induced cell injury. Taken together, these results demonstrate that PFOA induces cell injury through inhibition of autophagosomes formation and induction of ER stress in Leydig cells. Thus, our study sheds light on the cellular mechanisms of PFOA-induced Leydig cell injury, which may be suggestive to human male reproductive health risk assessment and prevention from PFOA exposure-induced reproductive toxicity.


Sujet(s)
Autophagosomes , Fluorocarbones , Cellules de Leydig , Souris , Animaux , Humains , Mâle , Stress du réticulum endoplasmique , Caprylates/toxicité , Apoptose
10.
Adv Biol (Weinh) ; 8(2): e2300477, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37867281

RÉSUMÉ

In recent years, there has been growing concern over the rising incidence of liver diseases, with increasing exposure to environmental toxins as a significant contributing factor. However, the mechanisms of liver injury induced by environmental pollutants are largely unclear. Here, using tetrabromobisphenol A (TBBPA), a widely used brominated flame retardant, as an example, environmental toxin-induced liver toxicity in mice is characterized via single-cell sequencing technology. Heterogeneous gene expression profiles after exposure to TBBPA in major cell types of the liver are demonstrated. In hepatocytes, pathway analysis of differentially expressed genes reveals the enhanced interferon response and diminished metabolic processes. The disrupted endothelial functions in TBBPA-treated cells are then shown. Moreover, the activation of M2-polarization in Kupffer cells, as well as activated effector T and B cells are unveiled in TBBPA-treated cells. Finally, ligand-receptor pair analysis shows that TBBPA disrupts cell-cell communication and induces an inflammatory microenvironment. Overall, the results reveal that TBBPA-induced dysfunction of hepatocytes and endothelial cells may then activate and recruit other immune cells such as Kuffer cells, and T/NK cells into the liver, further increasing inflammatory response and liver injury. Thus, the results provide novel insight into undesiring environmental pollutant-induced liver injury.


Sujet(s)
Polluants environnementaux , Polybromobiphényles , Souris , Animaux , Cellules endothéliales , Foie/métabolisme , Polybromobiphényles/toxicité , Polybromobiphényles/métabolisme , Polluants environnementaux/métabolisme , Analyse de séquence d'ARN
11.
Acta Pharmacol Sin ; 45(2): 391-404, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37803139

RÉSUMÉ

Hepatocellular carcinoma (HCC) is one of the most common and deadly cancers in the world. The therapeutic outlook for HCC patients has significantly improved with the advent and development of systematic and targeted therapies such as sorafenib and lenvatinib; however, the rise of drug resistance and the high mortality rate necessitate the continuous discovery of effective targeting agents. To discover novel anti-HCC compounds, we first constructed a deep learning-based chemical representation model to screen more than 6 million compounds in the ZINC15 drug-like library. We successfully identified LGOd1 as a novel anticancer agent with a characteristic levoglucosenone (LGO) scaffold. The mechanistic studies revealed that LGOd1 treatment leads to HCC cell death by interfering with cellular copper homeostasis, which is similar to a recently reported copper-dependent cell death named cuproptosis. While the prototypical cuproptosis is brought on by copper ionophore-induced copper overload, mechanistic studies indicated that LGOd1 does not act as a copper ionophore, but most likely by interacting with the copper chaperone protein CCS, thus LGOd1 represents a potentially new class of compounds with unique cuproptosis-inducing property. In summary, our findings highlight the critical role of bioavailable copper in the regulation of cell death and represent a novel route of cuproptosis induction.


Sujet(s)
Carcinome hépatocellulaire , Apprentissage profond , Tumeurs du foie , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Cuivre , Tumeurs du foie/traitement médicamenteux , Ionophores , Apoptose
12.
J Pharm Anal ; 13(8): 880-893, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-37719193

RÉSUMÉ

Triptolide is a key active component of the widely used traditional Chinese herb medicine Tripterygium wilfordii Hook. F. Although triptolide exerts multiple biological activities and shows promising efficacy in treating inflammatory-related diseases, its well-known safety issues, especially reproductive toxicity has aroused concerns. However, a comprehensive dissection of triptolide-associated testicular toxicity at single cell resolution is still lacking. Here, we observed testicular toxicity after 14 days of triptolide exposure, and then constructed a single-cell transcriptome map of 59,127 cells in mouse testes upon triptolide-treatment. We identified triptolide-associated shared and cell-type specific differentially expressed genes, enriched pathways, and ligand-receptor pairs in different cell types of mouse testes. In addition to the loss of germ cells, our results revealed increased macrophages and the inflammatory response in triptolide-treated mouse testes, suggesting a critical role of inflammation in triptolide-induced testicular injury. We also found increased reactive oxygen species (ROS) signaling and downregulated pathways associated with spermatid development in somatic cells, especially Leydig and Sertoli cells, in triptolide-treated mice, indicating that dysregulation of these signaling pathways may contribute to triptolide-induced testicular toxicity. Overall, our high-resolution single-cell landscape offers comprehensive information regarding triptolide-associated gene expression profiles in major cell types of mouse testes at single cell resolution, providing an invaluable resource for understanding the underlying mechanism of triptolide-associated testicular injury and additional discoveries of therapeutic targets of triptolide-induced male reproductive toxicity.

13.
Aging Dis ; 14(3): 652-669, 2023 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-37191408

RÉSUMÉ

A key pathological feature of neurodegenerative diseases (NDs) such as Alzheimer's disease (AD) and Parkinson's disease (PD) is the accumulation of aggregated and misfolded protein aggregates with limited effective therapeutic agents. TFEB (transcription factor EB), a key regulator of lysosomal biogenesis and autophagy, plays a pivotal role in the degradation of protein aggregates and has thus been regarded as a promising therapeutic target for these NDs. Here, we systematically summarize the molecular mechanisms and function of TFEB regulation. We then discuss the roles of TFEB and autophagy-lysosome pathways in major neurodegenerative diseases including AD and PD. Finally, we illustrate small molecule TFEB activators with protective roles in NDs animal models, which show great potential for being further developed into novel anti-neurodegenerative agents. Overall, targeting TFEB for enhancing lysosomal biogenesis and autophagy may represent a promising opportunity for the discovery of disease-modifying therapeutics for neurodegenerative disorders though more in-depth basic and clinical studies are required in the future.

15.
J Hazard Mater ; 448: 130785, 2023 04 15.
Article de Anglais | MEDLINE | ID: mdl-36860030

RÉSUMÉ

Tributyltin chloride (TBTCL), a commonly used antiseptic substance, is commonly found in the environment. Human exposure to TBTCL through the consumption of contaminated seafood, fish, or drinking water has aroused concern. It is well-characterized that TBTCL has multiple detrimental effects on the male reproductive system. However, the potential cellular mechanisms are not fully elucidated. Here, we characterized molecular mechanisms of TBTCL-induced cell injury in Leydig cells, a critical supporter for spermatogenesis. We showed that TBTCL induces apoptosis and cell cycle arrest in TM3 mouse Leydig cells. RNA sequencing analyses revealed that endoplasmic reticulum (ER) stress and autophagy were potentially involved in TBTCL-induced cytotoxicity. We further showed that TBTCL causes ER stress and inhibited autophagy flux. Notably, the inhibition of ER stress attenuates not only TBTCL-induces autophagy flux inhibition but also apoptosis and cell cycle arrest. Meanwhile, the activation of autophagy alleviates, and inhibition of autophagy exaggerates TBTCL-induced apoptosis and cell cycle arrest flux. These results suggest that TBTCL-induced ER stress and autophagy flux inhibition contributed to apoptosis and cell cycle arrest in Leydig cells, providing novel understanding into the mechanisms of TBTCL-induced testis toxicity.


Sujet(s)
Autophagie , Cellules de Leydig , Animaux , Humains , Souris , Mâle , Testicule , Stress du réticulum endoplasmique
17.
Autophagy ; 19(2): 706-715, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-35737739

RÉSUMÉ

Dysregulation of macroautophagy/autophagy has been closely implicated in aging. Caloric restriction (CR) is an effective intervention of aging partially via activation of autophagy. Recently, a high-throughput single-cell RNA-seq technique has been employed to detect the comprehensive transcriptomes of individual cells. However, the transcriptional networks of ATG (autophagy related) genes in the aging process and the modulation of ATG genes expression by CR at the single-cell level have not been elucidated. Here, by performing data analysis of single nucleus/cells RNA sequencing in rats undergoing aging and the modulation by CR, we demonstrate that the transcription patterns of Atg genes in different cell types of rat liver, brain, and kidney are highly heterogeneous. Importantly, CR reversed aging-induced changes of multiple Atg genes across different cell types in the brain, liver, and kidney. In summary, our results, for the first time, provide comprehensive information on Atg gene expression in specific cell types of different organs in a mammal during aging and give novel insight into the protective role of autophagy and CR in aging at the single-cell resolution.Abbreviations: ATG genes: autophagy-related genes; Atg5: autophagy related 5; Atg7: autophagy related 7; CR: caloric restriction; DEATG: differentially expressed autophagy-related; NAFLD: nonalcoholic fatty liver disease; ScRNA-seq: single-cell RNA sequencing.


Sujet(s)
Restriction calorique , Stéatose hépatique non alcoolique , Animaux , Rats , Vieillissement/génétique , Vieillissement/métabolisme , Autophagie/physiologie , Encéphale/métabolisme , Rein/métabolisme , Mammifères/génétique , Analyse de séquence d'ARN , Noyau de la cellule/génétique , Noyau de la cellule/métabolisme
18.
J Adv Res ; 53: 219-234, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-36528294

RÉSUMÉ

INTRODUCTION: Advanced paternal age of reproduction is an increasing trend, especially in developed countries and areas. This trend results in elevated risks of adverse reproductive outcomes such as reduced fertility rates, increased pregnancy loss, and poor childhood health. Yet, a systematic profiling of aging-associated molecular and cellular alterations in testicular tissue is still missing. OBJECTIVES: We aimed to dissect aging-associated molecular characteristics in testes of mice. METHODS: Single-cell transcriptomic sequencing and analysis were conducted in testes of young (2 months old) and old mice (24 months old). Immunofluorescences and immunochemistry were used to characterize aging-associated phenotypes and verify single cell sequence results. RESULTS: Here, we constructed the first single-cell transcriptomic atlases of testes of young and old mice. In-depth dissection of aging-dependent transcriptional alterations in specific cell types revealed multiple dysregulated biological processes such as increased 'senescence-associated secretory phenotype' and 'inflammation', which were major aging-associated characteristics. Further analysis of aging-related differentially expressed genes uncovered a disrupted balance of undifferentiated and differentiated spermatogonia stem cells in spermatogonia, indicative of a potential role of spermatogonia stem cells in aging-associated subfertility. Importantly, for the first time, our results identified an increased subtype of aging-specific macrophages, which may contribute to a hostile proinflammatory microenvironment during testicular aging. CONCLUSION: Taken together, our findings depict the distinct single-cell transcriptional features of the aged mouse testes and provide enormous resources for a comprehensive understanding of the cell-type-specific molecular mechanisms underlying mouse testicular aging, which may shed light on developing novel potential diagnostic biomarkers and therapeutic targets for age-associated male subfertility.


Sujet(s)
Infertilité masculine , Transcriptome , Humains , Grossesse , Femelle , Mâle , Souris , Animaux , Enfant , Nourrisson , Enfant d'âge préscolaire , Testicule/métabolisme , Spermatogonies/métabolisme , Vieillissement/génétique , Infertilité masculine/métabolisme
19.
Biomater Res ; 26(1): 64, 2022 Nov 26.
Article de Anglais | MEDLINE | ID: mdl-36435809

RÉSUMÉ

BACKGROUND: It is highly desirable to develop new therapeutic strategies for gastric cancer given the low survival rate despite improvement in the past decades. Cadherin 17 (CDH17) is a membrane protein highly expressed in cancers of digestive system. Nanobody represents a novel antibody format for cancer targeted imaging and drug delivery. Nanobody targeting CHD17 as an imaging probe and a delivery vehicle of toxin remains to be explored for its theragnostic potential in gastric cancer. METHODS: Naïve nanobody phage library was screened against CDH17 Domain 1-3 and identified nanobodies were extensively characterized with various assays. Nanobodies labeled with imaging probe were tested in vitro and in vivo for gastric cancer detection. A CDH17 Nanobody fused with toxin PE38 was evaluated for gastric cancer inhibition in vitro and in vivo. RESULTS: Two nanobodies (A1 and E8) against human CDH17 with high affinity and high specificity were successfully obtained. These nanobodies could specifically bind to CDH17 protein and CDH17-positive gastric cancer cells. E8 nanobody as a lead was extensively determined for tumor imaging and drug delivery. It could efficiently co-localize with CDH17-positive gastric cancer cells in zebrafish embryos and rapidly visualize the tumor mass in mice within 3 h when conjugated with imaging dyes. E8 nanobody fused with toxin PE38 showed excellent anti-tumor effect and remarkably improved the mice survival in cell-derived (CDX) and patient-derived xenograft (PDX) models. The immunotoxin also enhanced the anti-tumor effect of clinical drug 5-Fluorouracil. CONCLUSIONS: The study presents a novel imaging and drug delivery strategy by targeting CDH17. CDH17 nanobody-based immunotoxin is potentially a promising therapeutic modality for clinical translation against gastric cancer.

20.
Precis Clin Med ; 5(4): pbac023, 2022 Dec.
Article de Anglais | MEDLINE | ID: mdl-36349141

RÉSUMÉ

Background: Aristolochic acids (AAs), a class of carcinogenic and mutagenic natural products from Aristolochia and Asarum plants, are well-known to be responsible for inducing nephrotoxicity and urothelial carcinoma. Recently, accumulating evidence suggests that exposure to AAs could also induce hepatotoxicity and even hepatocellular carcinoma, though the mechanisms are poorly defined. Methods: Here, we aimed to dissect the underlying cellular and molecular mechanisms of aristolochic acid I (AAI)-induced hepatotoxicity by using advanced single-cell RNA sequencing (scRNA-seq) and proteomics techniques. We established the first single-cell atlas of mouse livers in response to AAI. Results: In hepatocytes, our results indicated that AAI activated NF-κB and STAT3 signaling pathways, which may contribute to the inflammatory response and apoptosis. In liver sinusoidal endothelial cells (LSECs), AAI activated multiple oxidative stress and inflammatory associated signaling pathways and induced apoptosis. Importantly, AAI induced infiltration of cytotoxic T cells and activation of proinflammatory macrophage and neutrophil cells in the liver to produce inflammatory cytokines to aggravate inflammation. Conclusions: Collectively, our study provides novel knowledge of AAs-induced molecular characteristics of hepatotoxicity at a single-cell level and suggests future treatment options for AAs associated hepatotoxicity.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE