Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22.978
Filtrer
1.
Biomaterials ; 313: 122796, 2025 Feb.
Article de Anglais | MEDLINE | ID: mdl-39226654

RÉSUMÉ

Chemotherapy-induced cellular senescence leads to an increased proportion of cancer stem cells (CSCs) in breast cancer (BC), contributing to recurrence and metastasis, while effective means to clear them are currently lacking. Herein, we aim to develop new approaches for selectively killing senescent-escape CSCs. High CD276 (95.60%) expression in multidrug-resistant BC cells, facilitates immune evasion by low-immunogenic senescent escape CSCs. CALD1, upregulated in ADR-resistant BC, promoting senescent-escape of CSCs with an anti-apoptosis state and upregulating CD276, PD-L1 to promote chemoresistance and immune escape. We have developed a controlled-released thermosensitive hydrogel containing pH- responsive anti-CD276 scFV engineered biomimetic nanovesicles to overcome BC in primary, recurrent, metastatic and abscopal humanized mice models. Nanovesicles coated anti-CD276 scFV selectively fuses with cell membrane of senescent-escape CSCs, then sequentially delivers siCALD1 and ADR due to pH-responsive MnP shell. siCALD1 together with ADR effectively induce apoptosis of CSCs, decrease expression of CD276 and PD-L1, and upregulate MHC I combined with Mn2+ to overcome chemoresistance and promote CD8+T cells infiltration. This combined therapeutic approach reveals insights into immune surveillance evasion by senescent-escape CSCs, offering a promising strategy to immunotherapy effectiveness in cancer therapy.


Sujet(s)
Tumeurs du sein , Vieillissement de la cellule , Résistance aux médicaments antinéoplasiques , Cellules souches tumorales , Humains , Animaux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/thérapie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Femelle , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Génie génétique/méthodes , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Nanoparticules/composition chimique , Anticorps à chaîne unique/composition chimique , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Antigène CD274/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Biomimétique/méthodes , Antigènes B7
2.
Sci Rep ; 14(1): 21852, 2024 09 19.
Article de Anglais | MEDLINE | ID: mdl-39300121

RÉSUMÉ

5-Fluorouracil (5-FU) is used as a standard first-line drug for colorectal cancer malignancy (CRC), but it brings a series of side effects such as severe diarrhea and intestinal damage. Our previous study found that a large number of senescent cells increased while 5-Fu induced intestinal damage, and anti-senescence drugs can alleviate its side effects of inflammatory damage. Oleanolic acid (OA) is a common pentacyclic triterpenoid mainly derived from food fungi and medicinal plants, and studies have shown that it mainly possesses hepatoprotective, enzyme-lowering, anti-inflammatory, and anti-tumor effects. But its role in senescence is still unclear. In the present study, we demonstrated for the first time that OA ameliorated 5-Fu-induced human umbilical vein endothelial cells (HUVECs) and human normal intestinal epithelial cells (NCM460) in a 5-Fu-induced cellular senescence model by decreasing the activity of SA-ß-gal-positive cells, and the expression of senescence-associated proteins (p16), senescence-associated genes (p53 and p21), and senescence-associated secretory phenotypes (SASPs: IL-1ß, IL-6, IL-8, IFN-γ and TNF-α). Meanwhile, in this study, in a BALB/c mouse model, we demonstrated that 5-FU induced intestinal inflammatory response and injury, which was also found to be closely related to the increase of senescent cells, and that OA treatment was effective in ameliorating these adverse phenomena. Furthermore, our in vivo and in vitro studies showed that OA could alleviate senescence by inhibiting mTOR. In colon cancer cell models, OA also enhanced the ability of 5-FU to kill HCT116 cells and SW480 cells. Overall, this study demonstrates for the first time the potential role of OA in counteracting the side effects of 5-FU chemotherapy, providing a new option for the treatment of colorectal cancer to progressively achieve the goal of high efficacy and low toxicity of chemotherapy.


Sujet(s)
Vieillissement de la cellule , Fluorouracil , Cellules endothéliales de la veine ombilicale humaine , Inflammation , Acide oléanolique , Acide oléanolique/pharmacologie , Fluorouracil/effets indésirables , Fluorouracil/pharmacologie , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Animaux , Souris , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Inflammation/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Souris de lignée BALB C , Intestins/effets des médicaments et des substances chimiques , Intestins/anatomopathologie , Mâle , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie
3.
Biomed Pharmacother ; 179: 117433, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39260327

RÉSUMÉ

Anti-aging immunity induced by vaccines was recently reported to enable the elimination of senescent cells. However, the initial immune response to vaccination declines with age, and there is evidence that elderly dendritic cells (DCs) have a reduced capacity to stimulate T cells. Identification of alternative anti-aging vaccine is therefore warranted. Here, we developed a DC vaccine that delivers a cationic protein (CP) fused with the seno-antigen peptides Gpnmb (Gpnmb-CP) into DCs. The Gpnmb-CP-pulsed DC vaccine (Gpnmb-CP-DC) efficiently presented antigens and activated CD8+ T cells, leading to enhanced immune cytotoxicity and memory responses in CD8+ T cells. Thus, the targeted anti-aging immunity triggered by Gpnmb-CP-DC has the ability to selectively eliminate senescent adipocytes and effectively improve age-related metabolic abnormalities in both high-fat diet (HFD)-induced young and aged mice models, as well as in natural aging mouse model. In contrast, the Gpnmb-CP protein vaccine exhibits minimal efficacy in aged mice model. Furthermore, we observed a decreased phagocytic capacity for antigens in aging DCs, accompanied by an upregulation of the immune checkpoint PDL1 expression and a noticeable decline in activated CD8+ T cell. Hence, Gpnmb-CP-DC emerges as a promising vaccine candidate, demonstrating the capacity to induce potent anti-aging immunity, mitigating adipose tissue senescence and metabolic abnormalities, while resilient to the senescent environment of the organism.


Sujet(s)
Tissu adipeux , Vieillissement , Lymphocytes T CD8+ , Cellules dendritiques , Souris de lignée C57BL , Animaux , Cellules dendritiques/immunologie , Vieillissement/immunologie , Souris , Lymphocytes T CD8+/immunologie , Tissu adipeux/immunologie , Tissu adipeux/métabolisme , Vieillissement de la cellule/immunologie , Vaccins/immunologie , Mâle , Alimentation riche en graisse , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/immunologie
4.
Theranostics ; 14(14): 5429-5442, 2024.
Article de Anglais | MEDLINE | ID: mdl-39310100

RÉSUMÉ

Background: Diabetic foot ulcers (DFUs) pose a substantial healthcare challenge due to their high rates of morbidity, recurrence, disability, and mortality. Current DFU therapeutics continue to grapple with multiple limitations. Senescent cells (SnCs) have been found to have a beneficial effect on acute wound healing, however, their roles in chronic wounds, such as DFU, remain unclear. Methods and results: We collected skin, fat, and muscle samples from clinical patients with DFU and lower limb fractures. RNA-sequencing combined with qPCR analyses on these samples demonstrate a significant accumulation of SnCs at DFU, as indicated by higher senescence markers (e.g., p16 and p21) and a senescence-associated secretory phenotype (SASP). We constructed a type 2 diabetic model of db/db mice, fed with a high-fat diet (Db-HFD), which were wounded using a 6 mm punch to the dorsal skin. HFD slightly affected wound healing in wild-type (WT) mice, but high glucose significantly delayed wound healing in the Db-HFD mice. We injected the mice with a previously developed fluorescent probe (XZ1208), which allows the detection of SnCs in vivo, and observed a strong senescence signal at the wound site of the Db-HFD mice. Contrary to the beneficial effects of SnCs in acute wound healing, our results demonstrated that clearance of SnCs using the senolytic compound ABT263 significantly accelerated wound healing in Db-HFD mice. Conclusion: Collectively, these findings suggest that SnCs critically accumulate at wound sites, delaying the healing process in DFUs. Thus, targeting SnCs with senolytic therapy represents a promising approach for DFU treatment, potentially improving the quality of life for patients with DFUs.


Sujet(s)
Vieillissement de la cellule , Diabète de type 2 , Pied diabétique , Peau , Cicatrisation de plaie , Animaux , Souris , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Diabète de type 2/complications , Humains , Pied diabétique/thérapie , Pied diabétique/métabolisme , Mâle , Peau/anatomopathologie , Peau/métabolisme , Alimentation riche en graisse/effets indésirables , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Souris de lignée C57BL , Modèles animaux de maladie humaine , Diabète expérimental , Phénotype sécrétoire associé à la sénescence , Femelle , Sulfonamides
5.
Clin Transl Med ; 14(9): e1772, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39270064

RÉSUMÉ

Tumour cell senescence can be induced by various factors, including DNA damage, inflammatory signals, genetic toxins, ionising radiation and nutrient metabolism. The senescence-associated secretory phenotype (SASP), secreted by senescent tumour cells, possesses the capacity to modulate various immune cells, including macrophages, T cells, natural killer cells and myeloid-derived suppressor cells, as well as vascular endothelial cells and fibroblasts within the tumour microenvironment (TME), and this modulation can result in either the promotion or suppression of tumorigenesis and progression. Exploring the impact of SASP on the TME could identify potential therapeutic targets, yet limited studies have dissected its functions. In this review, we delve into the causes and mechanisms of tumour cell senescence. We then concentrate on the influence of SASP on the tumour immune microenvironment, angiogenesis, extracellular matrix and the reprogramming of cancer stem cells, along with their associated tumour outcomes. Last, we present a comprehensive overview of the diverse array of senotherapeutics, highlighting their prospective advantages and challenge for the treatment of cancer patients. KEY POINTS: Senescence-associated secretory phenotype (SASP) secretion from senescent tumour cells significantly impacts cancer progression and biology. SASP is involved in regulating the remodelling of the tumour microenvironment, including immune microenvironment, vascular, extracellular matrix and cancer stem cells. Senotherapeutics, such as senolytic, senomorphic, nanotherapy and senolytic vaccines, hold promise for enhancing cancer treatment efficacy.


Sujet(s)
Tumeurs , Phénotype sécrétoire associé à la sénescence , Microenvironnement tumoral , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Humains , Tumeurs/immunologie , Tumeurs/métabolisme , Vieillissement de la cellule
6.
Biomater Adv ; 165: 214010, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39222592

RÉSUMÉ

The application of biomaterials in bone regeneration is a prevalent clinical practice. However, its efficacy in elderly patients remains suboptimal, necessitating further advancements. While biomaterial properties are known to orchestrate macrophage (MΦ) polarization and local immune responses, the role of biomaterial cues, specifically stiffness, in directing the senescent macrophage (S-MΦ) is still poorly understood. This study aimed to elucidate the role of substrate stiffness in modulating the immunomodulatory properties of S-MΦ and their role in osteo-immunomodulation. Our results demonstrated that employing collagen-coated polyacrylamide hydrogels with varying stiffness values (18, 76, and 295 kPa) as model materials, the high-stiffness hydrogel (295 kPa) steered S-MΦs towards a pro-inflammatory M1 phenotype, while hydrogels with lower stiffness (18 and 76 kPa) promoted an anti-inflammatory M2 phenotype. The immune microenvironment created by S-MΦs promoted the bioactivities of senescent endothelial cells (S-ECs) and senescent bone marrow mesenchymal stem cells BMSCs (S-BMSCs). Furthermore, the M2 S-MΦs, particularly incubated on the 76 kPa hydrogel matrices, significantly enhanced the ability of angiogenesis of S-ECs and osteogenic differentiation of S-BMSCs, which are crucial and interrelated processes in bone healing. This modulation aided in reducing the accumulation of reactive oxygen species in S-ECs and S-BMSCs, thereby significantly contributing to the repair and regeneration of aged bone tissue.


Sujet(s)
Régénération osseuse , Hydrogels , Immunomodulation , Macrophages , Cellules souches mésenchymateuses , Ostéogenèse , Régénération osseuse/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Hydrogels/composition chimique , Ostéogenèse/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/immunologie , Animaux , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Humains , Différenciation cellulaire , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Résines acryliques/composition chimique , Résines acryliques/pharmacologie , Matériaux biocompatibles/pharmacologie , Propriétés de surface , Collagène/métabolisme
7.
Cells ; 13(17)2024 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-39272993

RÉSUMÉ

Ovarian aging results in reproductive disorders and infertility in mammals. Previous studies have reported that the ferroptosis and autophagy caused by oxidative stress may lead to ovarian aging, but the mechanisms remain unclear. In this study, we compared the morphological characteristics between the aged and young ovaries of pigs and found that the aged ovaries were larger in size and showed more corpora lutea. TUNEL assay further showed that the apoptosis level of granulosa cells (GCs) was relatively higher in the aged ovaries than those in young ovaries, as well as the expressions of autophagy-associated genes, e.g., p62, ATG7, ATG5, and BECN1, but that the expressions of oxidative stress and aging-associated genes, e.g., SOD1, SIRT1, and SIRT6, were significantly lower. Furthermore, the RNA-seq, Western blotting, and immunofluorescence suggested that phospholipid phosphatase 3 (PLPP3) protein was significantly upregulated in the aged ovaries. PLPP3 was likely to decrease the expressions of SIRT1 and SIRT6 to accelerate cellular senescence of porcine GCs, inhibit the expressions of SOD1, CAT, FSP1, FTH1, and SLC7A11 to exacerbate oxidative stress and ferroptosis, and arouse autophagy to retard the follicular development. In addition, two SNPs of PLPP3 promoter were significantly associated with the age at puberty. g.155798586 (T/T) and g.155798718 (C/C) notably facilitated the mRNA and protein level of PLPP3. In conclusion, PLPP3 might aggravate the oxidative stress of GCs to accelerate ovarian aging, and two molecular markers of PLPP3 were identified for ovarian aging in pigs. This work not only contributes to investigations on mechanisms for ovarian aging but also provides valuable molecular markers to postpone ovarian aging in populations.


Sujet(s)
Vieillissement , Cellules de la granulosa , Ovaire , Stress oxydatif , Animaux , Femelle , Ovaire/métabolisme , Ovaire/anatomopathologie , Suidae , Vieillissement/génétique , Vieillissement/métabolisme , Cellules de la granulosa/métabolisme , Autophagie/génétique , Apoptose/génétique , Vieillissement de la cellule/génétique , Phosphatidate phosphatase/métabolisme , Phosphatidate phosphatase/génétique
8.
Cells ; 13(17)2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39273040

RÉSUMÉ

Aging is an inevitable biological process that contributes to the onset of age-related diseases, often as a result of mitochondrial dysfunction. Understanding the mechanisms behind aging is crucial for developing therapeutic interventions. This study investigates the effects of curcumin on postmitotic cellular lifespan (PoMiCL) during chronological aging in yeast, a widely used model for human postmitotic cellular aging. Our findings reveal that curcumin significantly prolongs the PoMiCL of wildtype yeast cells, with the most pronounced effects observed at lower concentrations, indicating a hormetic response. Importantly, curcumin also extends the lifespan of postmitotic cells with mitochondrial deficiencies, although the hormetic effect is absent in these defective cells. Mechanistically, curcumin inhibits TORC1 activity, enhances ATP levels, and induces oxidative stress. These results suggest that curcumin has the potential to modulate aging and offer therapeutic insights into age-related diseases, highlighting the importance of context in its effects.


Sujet(s)
Curcumine , Mitochondries , Saccharomyces cerevisiae , Curcumine/pharmacologie , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Saccharomyces cerevisiae/effets des médicaments et des substances chimiques , Saccharomyces cerevisiae/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Adénosine triphosphate/métabolisme , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Facteurs de transcription
9.
Cells ; 13(17)2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39273039

RÉSUMÉ

NAD+-dependent deacetylase sirtuin-1 (Sirt1) belongs to the sirtuins family, known to be longevity regulators, and exerts a key role in the prevention of vascular aging. By aging, the expression levels of Sirt1 decline with a severe impact on vascular function, such as the rise of endothelial dysfunction, which in turn promotes the development of cardiovascular diseases. In this context, the impact of Sirt1 activity in preventing endothelial senescence is particularly important. Given the key role of Sirt1 in counteracting endothelial senescence, great efforts have been made to deepen the knowledge about the intricate cross-talks and interactions of Sirt1 with other molecules, in order to set up possible strategies to boost Sirt1 activity to prevent or treat vascular aging. The aim of this review is to provide a proper background on the regulation and function of Sirt1 in the vascular endothelium and to discuss the recent advances regarding the therapeutic strategies of targeting Sirt1 to counteract vascular aging.


Sujet(s)
Vieillissement , Endothélium vasculaire , Sirtuine-1 , Humains , Sirtuine-1/métabolisme , Vieillissement/métabolisme , Animaux , Endothélium vasculaire/métabolisme , Vieillissement de la cellule , Cellules endothéliales/métabolisme
10.
Int J Mol Sci ; 25(17)2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39273228

RÉSUMÉ

Vascular aging is an important factor leading to cardiovascular diseases such as hypertension and atherosclerosis. Hyperlipidemia or fat accumulation may play an important role in vascular aging and cardiovascular disease. Isopropyl 3-(3,4-dihydroxyphenyl)-2-hydroxypropanoate (IDHP) has biological activity and can exert cardiovascular protection, which may be related to ferroptosis. However, the exact mechanism remains undefined. We hypothesized that IDHP may have a protective effect on blood vessels by regulating vascular aging caused by hyperlipidemia or vascular wall fat accumulation. The aim of this study is to investigate the protective effect and mechanism of IDHP on palmitic acid-induced human umbilical vein endothelial cells (HUVEC) based on senescence and ferroptosis. We found that IDHP could delay vascular aging, reduce the degree of ferrous ion accumulation and lipid peroxidation, and protect vascular cells from injury. These effects may be achieved by attenuating excessive reactive oxygen species (ROS) and ferroptosis signaling pathways generated in vascular endothelial cells. In short, our study identified IDHP as one of the antioxidant agents to slow down lipotoxicity-induced vascular senescence through the ROS/ferroptosis pathway. IDHP has new medicinal value and provides a new therapeutic idea for delaying vascular aging in patients with dyslipidemia.


Sujet(s)
Vieillissement de la cellule , Ferroptose , Cellules endothéliales de la veine ombilicale humaine , Acide palmitique , Espèces réactives de l'oxygène , Transduction du signal , Humains , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Acide palmitique/pharmacologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Ferroptose/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Peroxydation lipidique/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie
11.
Int J Mol Sci ; 25(17)2024 Sep 07.
Article de Anglais | MEDLINE | ID: mdl-39273652

RÉSUMÉ

Aging, a complex process marked by molecular and cellular changes, inevitably influences tissue and organ homeostasis and leads to an increased onset or progression of many chronic diseases and conditions, one of which is age-related hearing loss (ARHL). ARHL, known as presbycusis, is characterized by the gradual and irreversible decline in auditory sensitivity, accompanied by the loss of auditory sensory cells and neurons, and the decline in auditory processing abilities associated with aging. The extended human lifespan achieved by modern medicine simultaneously exposes a rising prevalence of age-related conditions, with ARHL being one of the most significant. While our understanding of the molecular basis for aging has increased over the past three decades, a further understanding of the interrelationship between the key pathways controlling the aging process and the development of ARHL is needed to identify novel targets for the treatment of AHRL. The dysregulation of molecular pathways (AMPK, mTOR, insulin/IGF-1, and sirtuins) and cellular pathways (senescence, autophagy, and oxidative stress) have been shown to contribute to ARHL. However, the mechanistic basis for these pathways in the initiation and progression of ARHL needs to be clarified. Therefore, understanding how longevity pathways are associated with ARHL will directly influence the development of therapeutic strategies to treat or prevent ARHL. This review explores our current understanding of the molecular and cellular mechanisms of aging and hearing loss and their potential to provide new approaches for early diagnosis, prevention, and treatment of ARHL.


Sujet(s)
Vieillissement , Vieillissement de la cellule , Presbyacousie , Humains , Vieillissement/métabolisme , Animaux , Presbyacousie/métabolisme , Presbyacousie/génétique , Presbyacousie/anatomopathologie , Transduction du signal , Stress oxydatif , Perte d'audition/métabolisme , Perte d'audition/génétique , Perte d'audition/anatomopathologie , Autophagie , Sérine-thréonine kinases TOR/métabolisme
12.
Int J Mol Sci ; 25(17)2024 Sep 07.
Article de Anglais | MEDLINE | ID: mdl-39273655

RÉSUMÉ

Cellular senescence is a permanent condition of cell cycle arrest caused by a progressive shortening of telomeres defined as replicative senescence. Stem cells may also undergo an accelerated senescence response known as premature senescence, distinct from telomere shortening, as a response to different stress agents. Various treatment protocols have been developed based on epigenetic changes in cells throughout senescence, using different drugs and antioxidants, senolytic vaccines, or the reprogramming of somatic senescent cells using Yamanaka factors. Even with all the recent advancements, it is still unknown how different epigenetic modifications interact with genetic profiles and how other factors such as microbiota physiological conditions, psychological states, and diet influence the interaction between genetic and epigenetic pathways. The aim of this review is to highlight the new epigenetic modifications that are involved in stem cell senescence. Here, we review recent senescence-related epigenetic alterations such as DNA methylation, chromatin remodeling, histone modification, RNA modification, and non-coding RNA regulation outlining new possible targets for the therapy of aging-related diseases. The advantages and disadvantages of the animal models used in the study of cellular senescence are also briefly presented.


Sujet(s)
Vieillissement de la cellule , Méthylation de l'ADN , Épigenèse génétique , Cellules souches , Vieillissement de la cellule/génétique , Humains , Animaux , Cellules souches/métabolisme , Assemblage et désassemblage de la chromatine
13.
Sci Transl Med ; 16(764): eadg1777, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39259812

RÉSUMÉ

Aging is a complex multifactorial process associated with epigenome dysregulation, increased cellular senescence, and decreased rejuvenation capacity. Short-term cyclic expression of octamer-binding transcription factor 4 (Oct4), sex-determining region Y-box 2 (Sox2), Kruppel-like factor 4 (Klf4), and cellular myelocytomatosis oncogene (cMyc) (OSKM) in wild-type mice improves health but fails to distinguish cell states, posing risks to healthy cells. Here, we delivered a single dose of adeno-associated viruses (AAVs) harboring OSK under the control of the cyclin-dependent kinase inhibitor 2a (Cdkn2a) promoter to specifically partially reprogram aged and stressed cells in a mouse model of Hutchinson-Gilford progeria syndrome (HGPS). Mice showed reduced expression of proinflammatory cytokines and extended life spans upon aged cell-specific OSK expression. The bone marrow and spleen, in particular, showed pronounced gene expression changes, and partial reprogramming in aged HGPS mice led to a shift in the cellular composition of the hematopoietic stem cell compartment toward that of young mice. Administration of AAVs carrying Cdkn2a-OSK to naturally aged wild-type mice also delayed aging phenotypes and extended life spans without altering the incidence of tumor development. Furthermore, intradermal injection of AAVs carrying Cdkn2a-OSK led to improved wound healing in aged wild-type mice. Expression of CDKN2A-OSK in aging or stressed human primary fibroblasts led to reduced expression of inflammation-related genes but did not alter the expression of cell cycle-related genes. This targeted partial reprogramming approach may therefore facilitate the development of strategies to improve health and life span and enhance resilience in the elderly.


Sujet(s)
Vieillissement , Reprogrammation cellulaire , Vieillissement de la cellule , Inhibiteur p16 de kinase cycline-dépendante , Modèles animaux de maladie humaine , Facteur-4 de type Kruppel , Animaux , Facteur-4 de type Kruppel/métabolisme , Vieillissement/métabolisme , Souris , Humains , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Inhibiteur p16 de kinase cycline-dépendante/génétique , Marqueurs biologiques/métabolisme , Progeria/métabolisme , Progeria/génétique , Progeria/anatomopathologie , Dependovirus/métabolisme , Régions promotrices (génétique)/génétique
14.
BMC Med ; 22(1): 376, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39256772

RÉSUMÉ

BACKGROUND: Neurogenic erectile dysfunction, characterized by neurological repair disorders and progressive corpus cavernosum fibrosis (CCF), is an unbearable disease with limited treatment success. IL-17A exhibits a complex role in tissue remodelling. Nevertheless, the precise role and underlying mechanisms of IL-17A in CCF under denervation remain unclear. METHODS: PCR array was employed to identified differentially expressed genes between neurogenic ED and normal rats. IL-17A expression and its main target cells were analyzed using Western blotting, immunofluorescence and immunohistochemistry. The phenotypic regulation of IL-17A on corpus cavernosum smooth muscle cells (CSMCs) was evaluated by cell cycle experiments and SA-ß-Gal staining. The mechanism of IL-17A was elucidated using non-target metabolomics and siRNA technique. Finally, IL-17A antagonist and ABT-263 (an inhibitor of B-cell lymphoma 2/w/xL) were utilized to enhance the therapeutic effect in a rat model of neurogenic ED. RESULTS: IL-17A emerged as the most significantly upregulated gene in the corpus cavernosum of model rats. It augmented the senescence transformation and fibrotic response of CSMCs, and exhibited a strong correlation with CCF. Mechanistically, IL-17A facilitated CCF by activating the mTORC2-ACACA signalling pathway, upregulating of CSMCs lipid synthesis and senescence transition, and increasing the secretion of fibro-matrix proteins. In vivo, the blockade of IL-17A-senescence signalling improved erectile function and alleviated CCF in neurogenic ED. CONCLUSIONS: IL-17A assumes a pivotal role in denervated CCF by activating the mTORC2-ACACA signalling pathway, presenting itself as a potential therapeutic target for effectively overcoming CCF and erection rehabilitation in neurogenic ED.


Sujet(s)
Dysfonctionnement érectile , Fibrose , Interleukine-17 , Pénis , Transduction du signal , Animaux , Mâle , Dysfonctionnement érectile/traitement médicamenteux , Interleukine-17/métabolisme , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Pénis/innervation , Pénis/anatomopathologie , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Rat Sprague-Dawley , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
15.
Nat Commun ; 15(1): 7712, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39231947

RÉSUMÉ

Osteoarthritis is a degenerative joint disease with joint pain as the main symptom, caused by fibrosis and loss of articular cartilage. Due to the complexity and heterogeneity of osteoarthritis, there is a lack of effective individualized disease-modifying osteoarthritis drugs in clinical practice. Chondrocyte senescence is reported to participate in occurrence and progression of osteoarthritis. Here we show that small molecule 10-hydroxy-2-decenoic acid suppresses cartilage degeneration and relieves pain in the chondrocytes, cartilage explants from osteoarthritis patients, surgery-induced medial meniscus destabilization or naturally aged male mice. We further confirm that 10-hydroxy-2-decenoic acid exerts a protective effect by targeting the glycosylation site in the Asp_Arg_Hydrox domain of aspartyl ß-hydroxylase. Mechanistically, 10-hydroxy-2-decenoic acid alleviate cellular senescence through the ERK/p53/p21 and GSK3ß/p16 pathways in the chondrocytes. Our study uncovers that 10-hydroxy-2-decenoic acid modulate cartilage metabolism by targeting aspartyl ß-hydroxylase to inhibit chondrocyte senescence in osteoarthritis. 10-hydroxy-2-decenoic acid may be a promising therapeutic drug against osteoarthritis.


Sujet(s)
Cartilage articulaire , Vieillissement de la cellule , Chondrocytes , Acides gras monoinsaturés , Arthrose , Animaux , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Mâle , Arthrose/métabolisme , Arthrose/anatomopathologie , Arthrose/traitement médicamenteux , Arthrose/prévention et contrôle , Souris , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Humains , Acides gras monoinsaturés/pharmacologie , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Souris de lignée C57BL , Modèles animaux de maladie humaine , Femelle
16.
Phytomedicine ; 134: 155982, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39244941

RÉSUMÉ

BACKGROUND: Aging-induced decline in ciliary muscle function is an important factor in visual accommodative deficits in elderly adults. With this study, we provide an innovative investigation of the interaction between ciliary muscle aging and oxidative stress. METHODS: Tricolor guinea pigs were used for the experiments in vivo and primary guinea pig ciliary smooth muscle cells were used for the experiments in vitro. RESULTS: We enriched for genes associated with muscle-aging-lutein relationship using bioinformatics, including Nuclear factor-erythroid 2-related factor-2 (Nrf2), Glutathione Peroxidase (GPx) gene family, Superoxide Dismutase (SOD) gene family, NAD(P)H: Quinone Oxidoreductase 1 (NQO1) and Heme Oxygenase-1 (HO-1). After gavage to aged guinea pigs, lutein reduced Reactive Oxygen Species (ROS) and P21 levels in senescent ciliary muscle; lutein decreased refractive error and restored accommodation of the eye. In addition, lutein increased GPx, SOD, and Catalase (CAT) levels in serum; lutein increased GPx and CAT levels in ciliary bodies. Lutein regulated the expression of proteins such as Nrf2, Kelch-like ECH-associated protein 1 (Keap1), and downstream proteins in senescent ciliary bodies. Similarly, guinea pig ciliary muscle cell senescence was associated with oxidative stress. In vitro, 100 µM lutein reversed the damage caused by 800 µM H2O2; it reduced Senescence-Associated ß-galactosidase (SA-ß-Gal) and ROS activites, cell apoptosis and cell migration. Also, lutein increased the expression of smooth muscle contractile proteins. Lutein also increased the expression of Nrf2, GPx2, NQO1 and HO-1, decreased the expression of Keap1. A reduction in Nrf2 activity led to a reduction in the ability of lutein to activate antioxidant enzymes in the cells, thus reducing its inhibitory effect on cell senescence. CONCLUSION: lutein improved resistance to oxidative stress in senescent ciliary muscle in vivo and in vitro by regulating the Keap1/Nrf2/Antioxidant Response Element pathway. We have innovatively demonstrated the molecular pharmacological mechanism by which lutein reverse age-related ciliary muscle systolic and diastolic deficits.


Sujet(s)
Protéine-1 de type kelch associée à ECH , Lutéine , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Animaux , Cochons d'Inde , Stress oxydatif/effets des médicaments et des substances chimiques , Facteur-2 apparenté à NF-E2/métabolisme , Protéine-1 de type kelch associée à ECH/métabolisme , Lutéine/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Corps ciliaire/effets des médicaments et des substances chimiques , Vieillissement/effets des médicaments et des substances chimiques , Éléments de réponse aux anti-oxydants/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques
17.
Nat Commun ; 15(1): 8171, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39289351

RÉSUMÉ

T cell senescence alters the homeostasis of distinct T cell populations and results in decayed adaptive immune protection in older individuals, but a link between aging and dynamic T cell clone changes has not been made. Here, using a newly developed computational framework, Repertoire Functional Units (RFU), we investigate over 6500 publicly available TCR repertoire sequencing samples from multiple human cohorts and identify age-associated RFUs consistently across different cohorts. Quantification of RFU reduction with aging reveals accelerated loss under immunosuppressive conditions. Systematic analysis of age-associated RFUs in clinical samples manifests a potential link between these RFUs and improved clinical outcomes, such as lower ICU admission and reduced risk of complications, during acute viral infections. Finally, patients receiving bone marrow transplantation show a secondary expansion of the age-associated clones upon stem cell transfer from younger donors. Together, our results suggest the existence of a 'TCR clock' that could reflect the immune functions in aging populations.


Sujet(s)
Vieillissement , Récepteurs aux antigènes des cellules T , Lymphocytes T , Humains , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/immunologie , Vieillissement/immunologie , Sujet âgé , Lymphocytes T/immunologie , Adulte d'âge moyen , Adulte , Transplantation de moelle osseuse , Mâle , Femelle , Vieillissement de la cellule/immunologie , Jeune adulte , Sujet âgé de 80 ans ou plus
18.
Nat Aging ; 4(9): 1194-1210, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39251866

RÉSUMÉ

Reproductive aging is a major cause of fertility decline, attributed to decreased oocyte quantity and developmental potential. A possible cause is aging of the surrounding follicular somatic cells that support oocyte growth and development by providing nutrients and regulatory factors. Here, by creating chimeric follicles, whereby an oocyte from one follicle was transplanted into and cultured within another follicle whose native oocyte was removed, we show that young oocytes cultured in aged follicles exhibited impeded meiotic maturation and developmental potential, whereas aged oocytes cultured within young follicles were significantly improved in rates of maturation, blastocyst formation and live birth after in vitro fertilization and embryo implantation. This rejuvenation of aged oocytes was associated with enhanced interaction with somatic cells, transcriptomic and metabolomic remodeling, improved mitochondrial function and higher fidelity of meiotic chromosome segregation. These findings provide the basis for a future follicular somatic cell-based therapy to treat female infertility.


Sujet(s)
Ovocytes , Follicule ovarique , Rajeunissement , Femelle , Animaux , Follicule ovarique/croissance et développement , Rajeunissement/physiologie , Souris , Fécondation in vitro/méthodes , Vieillissement de la cellule , Méiose , Microenvironnement cellulaire , Vieillissement/physiologie
19.
Cell Mol Life Sci ; 81(1): 407, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39287670

RÉSUMÉ

Extension of the replicative lifespan of primary cells can be achieved by activating human telomerase reverse transcriptase (hTERT) to maintain sufficient telomere lengths. In this work, we utilize CRISPR/dCas9-based epigenetic modifiers (p300 histone acetyltransferase and TET1 DNA demethylase) and transcriptional activators (VPH and VPR) to reactivate the endogenous TERT gene in unstimulated T cells in the peripheral blood mononuclear cells (PBMCs) by rewiring the epigenetic marks of the TERT promoter. Importantly, we have successfully expanded resting T cells and delayed their cellular senescence for at least three months through TERT reactivation, without affecting the expression of a T-cell marker (CD3) or inducing an accelerated cell division rate. We have also demonstrated the effectiveness of these CRISPR tools in HEK293FT and THP-1-derived macrophages. TERT reactivation and replicative senescence delay were achieved without inducing malignancy transformation, as shown in various cellular senescence assays, cell cycle state, proliferation rate, cell viability, and karyotype analyses. Our chromatin immunoprecipitation (ChIP)-qPCR data together with TERT mRNA and protein expression analyses confirmed the specificity of CRISPR-based transcription activators in modulating epigenetic marks of the TERT promoter, and induced telomerase expression. Therefore, the strategy of cell immortalization described here can be potentially adopted and generalized to delay cell death or even immortalize any other cell types.


Sujet(s)
Systèmes CRISPR-Cas , Vieillissement de la cellule , Épigenèse génétique , Régions promotrices (génétique) , Lymphocytes T , Telomerase , Humains , Telomerase/génétique , Telomerase/métabolisme , Systèmes CRISPR-Cas/génétique , Vieillissement de la cellule/génétique , Régions promotrices (génétique)/génétique , Lymphocytes T/métabolisme , Lymphocytes T/cytologie , Cellules HEK293 , Prolifération cellulaire/génétique
20.
Cell Death Dis ; 15(9): 680, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39289337

RÉSUMÉ

Iron overload and cellular senescence have been implicated in liver fibrosis, but their possible mechanistic connection has not been explored. To address this, we have delved into the role of iron and senescence in an experimental model of chronic liver injury, analyzing whether an iron chelator would prevent liver fibrosis by decreasing hepatocyte senescence. The model of carbon tetrachloride (CCl4) in mice was used as an experimental model of liver fibrosis. Results demonstrated that during the progression of liver fibrosis, accumulation of iron occurs, concomitant with the appearance of fibrotic areas and cells undergoing senescence. Isolated parenchymal hepatocytes from CCl4-treated mice present a gene transcriptomic signature compatible with iron accumulation and senescence, which correlates with induction of Reactive Oxygen Species (ROS)-related genes, activation of the Transforming Growth Factor-beta (TGF-ß) pathway and inhibition of oxidative metabolism. Analysis of the iron-related gene signature in a published single-cell RNA-seq dataset from CCl4-treated livers showed iron accumulation correlating with senescence in other non-parenchymal liver cells. Treatment with deferiprone, an iron chelator, attenuated iron accumulation, fibrosis and senescence, concomitant with relevant changes in the senescent-associated secretome (SASP), which switched toward a more anti-inflammatory profile of cytokines. In vitro experiments in human hepatocyte HH4 cells demonstrated that iron accumulates in response to a senescence-inducing reagent, doxorubicin, being deferiprone able to prevent senescence and SASP, attenuating growth arrest and cell death. However, deferiprone did not significantly affect senescence induced by two different agents (doxorubicin and deoxycholic acid) or activation markers in human hepatic stellate LX-2 cells. Transcriptomic data from patients with different etiologies demonstrated the relevance of iron accumulation in the progression of liver chronic damage and fibrosis, correlating with a SASP-related gene signature and pivotal hallmarks of fibrotic changes. Altogether, our study establishes iron accumulation as a clinically exploitable driver to attenuate pathological senescence in hepatocytes.


Sujet(s)
Vieillissement de la cellule , Agents chélateurs du fer , Cirrhose du foie , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Cirrhose du foie/traitement médicamenteux , Animaux , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Agents chélateurs du fer/pharmacologie , Humains , Souris , Mâle , Évolution de la maladie , Fer/métabolisme , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/anatomopathologie , Souris de lignée C57BL , Tétrachloro-méthane , Défériprone/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Modèles animaux de maladie humaine
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE