Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.282
Filtrer
1.
Nat Commun ; 15(1): 7791, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39242637

RÉSUMÉ

Multiple sclerosis (MS) is a debilitating demyelinating disease characterized by remyelination failure attributed to inadequate oligodendrocyte precursor cells (OPCs) differentiation and aberrant astrogliosis. A comprehensive cell atlas reanalysis of clinical specimens brings to light heightened clusterin (CLU) expression in a specific astrocyte subtype links to active lesions in MS patients. Our investigation reveals elevated astrocytic CLU levels in both active lesions of patient tissues and female murine MS models. CLU administration stimulates primary astrocyte proliferation while concurrently impeding astrocyte-mediated clearance of myelin debris. Intriguingly, CLU overload directly impedes OPC differentiation and induces OPCs and OLs apoptosis. Mechanistically, CLU suppresses PI3K-AKT signaling in primary OPCs via very low-density lipoprotein receptor. Pharmacological activation of AKT rescues the damage inflicted by excess CLU on OPCs and ameliorates demyelination in the corpus callosum. Furthermore, conditional knockout of CLU emerges as a promising intervention, showcasing improved remyelination processes and reduced severity in murine MS models.


Sujet(s)
Astrocytes , Clusterine , Maladies démyélinisantes , Modèles animaux de maladie humaine , Remyélinisation , Animaux , Femelle , Humains , Souris , Apoptose/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Clusterine/métabolisme , Clusterine/génétique , Corps calleux/métabolisme , Corps calleux/anatomopathologie , Maladies démyélinisantes/métabolisme , Maladies démyélinisantes/anatomopathologie , Souris de lignée C57BL , Souris knockout , Sclérose en plaques/métabolisme , Sclérose en plaques/anatomopathologie , Gaine de myéline/métabolisme , Précurseurs des oligodendrocytes/métabolisme , Précurseurs des oligodendrocytes/effets des médicaments et des substances chimiques , Oligodendroglie/métabolisme , Oligodendroglie/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Remyélinisation/effets des médicaments et des substances chimiques , Transduction du signal
2.
Stem Cell Res Ther ; 15(1): 300, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39272156

RÉSUMÉ

BACKGROUND: Emerging evidence has highlighted the therapeutic potential of human umbilical cord mesenchymal stem cells (UC-MSCs) in chemotherapy-induced premature ovarian failure (POF). This study was designed to investigate the appropriate timing and molecular mechanism of UC-MSCs treatment for chemotherapy-induced POF. METHODS: Ovarian structure and function of mice were assessed every 3 days after injections with cyclophosphamide (CTX) and busulfan (BUS). UC-MSCs and UC-MSCs-derived extracellular vesicles (EVs) were infused into mice via the tail vein, respectively. Ovarian function was analyzed by follicle counts, the serum levels of hormones and ovarian morphology. The apoptosis and proliferation of ovarian granulosa cells were analyzed in vitro and in vivo. Label-free quantitative proteomics was used to detect the differentially expressed proteins in UC-MSC-derived EVs. RESULTS: After CTX/BUS injection, we observed that the ovarian function of POF mice was significantly deteriorated on day 9 after CTX/BUS infusion. TUNEL assay indicated that the number of apoptotic cells in the ovaries of POF mice was significantly higher than that in normal mice on day 3 after CTX/BUS injection. Transplantation of UC-MSCs on day 6 after CTX/BUS injection significantly improved ovarian function, enhanced proliferation and inhibited apoptosis of ovarian granulosa cells, whereas the therapeutic effect of UC-MSCs transplantation decreased on day 9, or day 12 after CTX/BUS injection. Moreover, EVs derived from UC-MSCs exerted similar therapeutic effects on POF. UC-MSCs-derived EVs could activate the PI3K/AKT signaling pathway and reduce ovarian granulosa cell apoptosis. Quantitative proteomics analysis revealed that clusterin (CLU) was highly expressed in the EVs of UC-MSCs. The supplementation of CLU proteins prevented ovarian granulosa cells from chemotherapy-induced apoptosis. Further mechanistic analysis showed that CLU-knockdown blocked the PI3K/AKT signaling and reversed the protective effects of UC-MSCs-derived EVs. CONCLUSIONS: Administration of UC-MSCs and UC-MSCs-derived EVs on day 6 of CTX/BUS injection could effectively improve the ovarian function of POF mice. UC-MSCs-derived EVs carrying CLU promoted proliferation and inhibited apoptosis of ovarian granulosa cells through activating the PI3K/AKT pathway. This study identifies a previously unrecognized molecular mechanism of UC-MSCs-mediated protective effects on POF, which pave the way for the use of cell-free therapeutic approach for POF.


Sujet(s)
Vésicules extracellulaires , Cellules souches mésenchymateuses , Phosphatidylinositol 3-kinases , Insuffisance ovarienne primitive , Protéines proto-oncogènes c-akt , Transduction du signal , Cordon ombilical , Femelle , Animaux , Insuffisance ovarienne primitive/thérapie , Insuffisance ovarienne primitive/métabolisme , Insuffisance ovarienne primitive/induit chimiquement , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/transplantation , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Souris , Protéines proto-oncogènes c-akt/métabolisme , Humains , Phosphatidylinositol 3-kinases/métabolisme , Cordon ombilical/cytologie , Clusterine/métabolisme , Apoptose , Transplantation de cellules souches mésenchymateuses/méthodes , Ovaire/métabolisme , Cellules de la granulosa/métabolisme , Prolifération cellulaire , Busulfan/pharmacologie
3.
Medicine (Baltimore) ; 103(32): e39202, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39121317

RÉSUMÉ

Vancomycin, a first-line drug for treating methicillin-resistant Staphylococcus aureus infections, is associated with acute kidney injury (AKI). This study involved an evaluation of biomarkers for AKI detection and their comparison with traditional serum creatinine (SCr). We prospectively enrolled patients scheduled to receive intravenous vancomycin for methicillin-resistant S aureus infection. Blood samples for pharmacokinetic assessment and SCr and cystatin C (CysC) measurements were collected at baseline and on days 3, 7, and 10 from the initiation of vancomycin administration (day 1). Urinary biomarkers, including kidney injury molecule 1 (KIM-1), neutrophil gelatinase-associated lipocalin, and clusterin, were collected from days 1 to 7 and adjusted for urinary creatinine levels. The estimated glomerular filtration rate (eGFR) was calculated using the Chronic Kidney Disease Epidemiology Collaboration equation. Of the 42 patients, 6 experienced vancomycin-induced AKI. On day 7, the change from baseline eGFR using CysC (ΔeGFRCysC) showed a stronger correlation with vancomycin area under the curve (r = -0.634, P < .001) than that using SCr (ΔeGFRSCr; r = -0.437, P = .020). ΔeGFRSCr showed no significant correlation with vancomycin pharmacokinetic in patients with body mass index ≥23. The median (interquartile range) level of KIM-1 (µg/mg) was significantly higher in the AKI group (0.006 [0.005-0.008]) than in the non-AKI group (0.004 [0.001-0.005]) (P = .039, Mann-Whitney U test), with area under the receiver operating characteristic curve (95% confidence interval) of 0.788 (0.587-0.990). Serum CysC, particularly in overweight individuals or those with obesity, along with urinary KIM-1 are important predictors of vancomycin-induced AKI. These results may aid in selecting better biomarkers than traditional SCr for detecting vancomycin-induced AKI.


Sujet(s)
Atteinte rénale aigüe , Antibactériens , Marqueurs biologiques , Créatinine , Cystatine C , Récepteur cellulaire-1 du virus de l'hépatite A , Vancomycine , Humains , Vancomycine/effets indésirables , Vancomycine/pharmacocinétique , Vancomycine/administration et posologie , Vancomycine/sang , Marqueurs biologiques/urine , Marqueurs biologiques/sang , Atteinte rénale aigüe/induit chimiquement , Atteinte rénale aigüe/urine , Atteinte rénale aigüe/sang , Mâle , Femelle , Études prospectives , Adulte d'âge moyen , Antibactériens/effets indésirables , Antibactériens/pharmacocinétique , Antibactériens/administration et posologie , Sujet âgé , Récepteur cellulaire-1 du virus de l'hépatite A/analyse , Cystatine C/sang , Cystatine C/urine , Créatinine/sang , Créatinine/urine , Débit de filtration glomérulaire , Lipocaline-2/urine , Lipocaline-2/sang , Infections à staphylocoques/traitement médicamenteux , Staphylococcus aureus résistant à la méticilline , Clusterine/urine , Clusterine/sang
4.
Clin Transl Sci ; 17(7): e13881, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38982592

RÉSUMÉ

Chondrocyte apoptosis is recognized as one of the pathological features involved in cartilage degeneration driving the onset and progression of knee osteoarthritis (OA). This study aimed to determine the molecular mechanism underlying the effect of clusterin (CLU), anti-apoptotic molecule, in human knee OA chondrocytes. Primary knee OA chondrocytes were isolated from the cartilage of knee OA patients and divided into five groups: (1) the cells treated with interleukin (IL)-1ß, (2) CLU alone, (3) a combination of IL-1ß and CLU, (4) LY294002 (PI3K inhibitor) along with IL-1ß and CLU, and (5) the untreated cells. Production of apoptotic, inflammatory, anabolic, and catabolic mediators in knee OA chondrocytes was determined after treatment for 24 h. Our in vitro study uncovered that CLU significantly suppressed the production of inflammatory mediators [nitric oxide (NO), IL6, and tumor necrosis factor (TNF)-α] and apoptotic molecule (caspase-3, CASP3). CLU significantly upregulated messenger ribonucleic acid (mRNA) expressions of anabolic factors [SRY-box transcription factor-9 (SOX9) and aggrecan (ACAN)], but significantly downregulated mRNA expressions of IL6, nuclear factor kappa-B (NF-κB), CASP3, and matrix metalloproteinase-13 (MMP13). Anti-apoptotic and anti-inflammatory effects of CLU were mediated through activating PI3K/Akt signaling pathway. The findings suggest that CLU might have beneficial effects on knee OA chondrocytes by exerting anti-apoptotic and anti-inflammatory functions via PI3K/Akt pathway, making CLU a promising target for potential therapeutic interventions in knee OA.


Sujet(s)
Apoptose , Chondrocytes , Clusterine , Interleukine-1 bêta , Gonarthrose , Humains , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Gonarthrose/anatomopathologie , Gonarthrose/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Clusterine/métabolisme , Clusterine/génétique , Interleukine-1 bêta/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Cellules cultivées , Mâle , Adulte d'âge moyen , Sujet âgé , Inflammation/métabolisme , Inflammation/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Femelle , Phosphatidylinositol 3-kinases/métabolisme , Morpholines/pharmacologie , 4H-1-Benzopyran-4-ones/pharmacologie , Facteur de transcription SOX-9/métabolisme , Facteur de transcription SOX-9/génétique , Matrix Metalloproteinase 13/métabolisme , Médiateurs de l'inflammation/métabolisme , Monoxyde d'azote/métabolisme
5.
Methods Mol Biol ; 2816: 145-149, 2024.
Article de Anglais | MEDLINE | ID: mdl-38977596

RÉSUMÉ

Clusterin, also known as apolipoprotein J, is an ATP-independent holdase chaperone protein. Clusterin is involved in various functions including protein quality control and lipid transport. Though clusterin is secreted upon stress, the intracellular fate of clusterin after a stress response is not well understood. The protocol described here utilizes clusterin tagged to fluorescent proteins like green fluorescent protein and red fluorescent protein to understand the intracellular fate of clusterin.


Sujet(s)
Clusterine , Microscopie confocale , Clusterine/métabolisme , Humains , Microscopie confocale/méthodes , Protéines à fluorescence verte/métabolisme , Protéines à fluorescence verte/génétique , Protéines luminescentes/métabolisme , Protéines luminescentes/génétique , , Animaux
6.
Neuroreport ; 35(13): 857-867, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-38973492

RÉSUMÉ

Sepsis-associated encephalopathy (SAE) is a severe complication of sepsis, however, its exact mechanism remains unknown. This study aimed to evaluate whether clusterin is essential to the development of SAE during the aging process of astrocytes. In the study, septic mice were established with cecal ligation and puncture (CLP) and lipopolysaccharides were applied to astrocytes in vitro. Evan's blue dye was used in vivo to show blood-brain barrier (BBB) permeability. A morris water maze test was conducted to assess cognitive functions of the mice. Clusterin-knockout mice were used to examine the effect of clusterin on sepsis. The astrocytes were transfected with lentivirus expressing clusterin cDNA for clusterin overexpression or pYr-LV-clusterin small hairpin RNA for clusterin knockdown in vitro . The expression of clusterin, p-p53, p21, GDNF, and iNOS was detected. he CLP mice exhibited a higher clusterin expression in hippocampus tissue, aging astrocytes, lower GDNF expression and higher iNOS expression, accompanied with BBB damage and cognitive deficiency. Following clusterin knockout, this pathological process was further enhanced. In vitro , following lipopolysaccharides treatment, astrocytes exhibited increased clusterin, p-p53, p21, iNOS and decreased GDNF. Following clusterin knockdown, the cells exhibited a further increase in p-p53, p21, and iNOS and decrease in GDNF. Clusterin overexpression, however, helped inhibit astrocytes aging and neuroinflammation evidenced by decreased p-p53, p21, iNOS and increased GDNF. The present study has revealed that clusterin may exert its neuroprotective effect by preventing aging in astrocytes, suppressing the secretion of iNOS and promoting GNDF release.


Sujet(s)
Astrocytes , Barrière hémato-encéphalique , Clusterine , Dysfonctionnement cognitif , Souris knockout , Encéphalopathie associée au sepsis , Animaux , Clusterine/métabolisme , Astrocytes/métabolisme , Barrière hémato-encéphalique/métabolisme , Encéphalopathie associée au sepsis/métabolisme , Souris , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/étiologie , Mâle , Souris de lignée C57BL , Vieillissement de la cellule/physiologie , Lipopolysaccharides , Sepsie/complications , Sepsie/métabolisme , Hippocampe/métabolisme
7.
Alzheimers Res Ther ; 16(1): 169, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39069622

RÉSUMÉ

BACKGROUND: Cerebral amyloid angiopathy (CAA) is characterized by amyloid-ß (Aß) deposition in cerebral vessels, leading to lobar cerebral microbleeds (CMB) and intracerebral hemorrhages (ICH). Apolipoprotein J (ApoJ) is a multifunctional chaperone related to Aß aggregation and clearance. Our study investigated the vascular impact of chronic recombinant human Apolipoprotein J (rhApoJ) treatment in a transgenic mouse model of ß-amyloidosis with prominent CAA. METHODS: Twenty-month-old APP23 C57BL/6 mice received 25 doses of rhApoJ (1 mg/kg) (n = 9) or saline (n = 8) intraperitoneally for 13 weeks, while Wild-type (WT) mice received saline (n = 13). Postmortem brains underwent T2*-weighted magnetic resonance imaging (MRI) to detect hemorrhagic lesions. Aß levels and distribution, cerebral fibrinogen leakage, brain smooth muscle actin (sma), and plasma matrix metalloproteinases and inflammatory markers were analyzed after treatments. Additionally, plasma samples from 22 patients with lobar ICH were examined to determine the clinical relevance of the preclinical findings. RESULTS: rhApoJ-treated APP23 presented fewer cortical CMBs (50-300 µm diameter) (p = 0.012) and cortical larger hemorrhages (> 300 µm) (p = 0.002) than saline-treated mice, independently of Aß brain levels. MRI-detected hemorrhagic lesions correlated with fibrinogen cerebral extravasation (p = 0.011). Additionally, rhApoJ-treated mice presented higher number of sma-positive vessels than saline-treated mice (p = 0.038). In rhApoJ-treated mice, human ApoJ was detected in plasma and in occasional leptomeningeal vessels, but not in the parenchyma, suggesting that its mechanism of action operates through the periphery. The administration of rhApoJ induced an increase in plasma Groα (p = 0.035) and MIP-1α (p = 0.035) levels, while lower MMP-12 (p = 0.046) levels, compared to the saline-treated group. In acute lobar ICH patients, MMP-12 plasma levels correlated with larger hemorrhage volume (p = 0.040) and irregular ICH shape (p = 0.036). CONCLUSIONS: Chronic rhApoJ treatment in aged APP23 mice ameliorated CAA-related neurovascular damage by reducing the occurrence of CMB. We propose that rhApoJ may prevent blood-brain barrier (BBB) leakage and CMB appearance partly through circulating MMP-12 modulation.


Sujet(s)
Angiopathie amyloïde cérébrale , Hémorragie cérébrale , Modèles animaux de maladie humaine , Souris de lignée C57BL , Souris transgéniques , Animaux , Angiopathie amyloïde cérébrale/traitement médicamenteux , Humains , Hémorragie cérébrale/sang , Souris , Mâle , Femelle , Peptides bêta-amyloïdes , Encéphale/imagerie diagnostique , Encéphale/anatomopathologie , Encéphale/effets des médicaments et des substances chimiques , Sujet âgé , Imagerie par résonance magnétique , Protéines recombinantes/administration et posologie , Précurseur de la protéine bêta-amyloïde/génétique , Clusterine
8.
Mol Med ; 30(1): 100, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38992588

RÉSUMÉ

BACKGROUND: Diabetes mellitus (DM)-induced testicular damage is associated with sexual dysfunction and male infertility in DM patients. However, the pathogenesis of DM-induced testicular damage remains largely undefined. METHODS: A streptozotocin (STZ)-induced diabetic model and high glucose (HG)-treated in vitro diabetic model were established. The histological changes of testes were assessed by H&E staining. Serum testosterone, iron, MDA and GSH levels were detected using commercial kits. Cell viability and lipid peroxidation was monitored by MTT assay and BODIPY 581/591 C11 staining, respectively. qRT-PCR, immunohistochemistry (IHC) or Western blotting were employed to detect the levels of BRD7, Clusterin, EZH2 and AMPK signaling molecules. The associations among BRD7, EZH2 and DNMT3a were detected by co-IP, and the transcriptional regulation of Clusterin was monitored by methylation-specific PCR (MSP) and ChIP assay. RESULTS: Ferroptosis was associated with DM-induced testicular damage in STZ mice and HG-treated GC-1spg cells, and this was accompanied with the upregulation of BRD7. Knockdown of BRD7 suppressed HG-induced ferroptosis, as well as HG-induced Clusterin promoter methylation and HG-inactivated AMPK signaling in GC-1spg cells. Mechanistical studies revealed that BRD7 directly bound to EZH2 and regulated Clusterin promoter methylation via recruiting DNMT3a. Knockdown of Clusterin or inactivation of AMPK signaling reverses BRD7 silencing-suppressed ferroptosis in GC-1spg cells. In vivo findings showed that lack of BRD7 protected against diabetes-induced testicular damage and ferroptosis via increasing Clusterin expression and activating AMPK signaling. CONCLUSION: BRD7 suppressed Clusterin expression via modulating Clusterin promoter hypermethylation in an EZH2 dependent manner, thereby suppressing AMPK signaling to facilitate ferroptosis and induce diabetes-associated testicular damage.


Sujet(s)
AMP-Activated Protein Kinases , Clusterine , Méthylation de l'ADN , Diabète expérimental , Ferroptose , Régions promotrices (génétique) , Transduction du signal , Testicule , Animaux , Mâle , Souris , AMP-Activated Protein Kinases/métabolisme , Lignée cellulaire , Clusterine/génétique , Clusterine/métabolisme , Diabète expérimental/métabolisme , Diabète expérimental/génétique , Diabète expérimental/complications , DNA methyltransferase 3A/métabolisme , Protéine-2 homologue de l'activateur de Zeste/génétique , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Ferroptose/génétique , Souris de lignée C57BL , Testicule/métabolisme , Testicule/anatomopathologie
9.
Mol Reprod Dev ; 91(7): e23764, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39072963

RÉSUMÉ

Clusterin (CLU), one of the main glycoproteins in mammalian semen and the male reproductive tract, plays a role in spermatogenesis and sperm maturation. Given the poor reliability of classic seminal studies in determining male-fertilizing capacity and the differences in CLU abundance between normal and abnormal spermatozoa, we investigated the potential value of mRNA-CLU levels and protein distribution in spermatozoa as markers of sperm quality and predictors of male fertility. This multicenter study included 90 patients undergoing in vitro fertilization (IVF) treatment with their partners, and a control group of 36 fertile males with normal seminograms. We assessed the relationship between IVF treatment outcomes, seminogram variables, mRNA-CLU levels by quantitative real-time-PCR and CLU distribution by immunostaining in spermatozoa. Our study reveals CLU staining in the acrosome (p = 0.002, OR 14.8, 95% CI: 2.7-79.3) and mRNA-CLU levels (p = 0.005, OR 10.85, 95% CI: 2.0-57.4) as independent risk factors for pregnancy failure, irrespective of traditional seminogram variables. Additionally, our results suggest that CLU, and specially its secreted isoform, constitutes a component of the protein pool that human spermatozoa can produce during its maturation process, exhibiting a variable abundance and distribution in spermatozoa from fertile men compared to those in patients with altered seminograms and infertile patients with normal seminograms. Our study is the first to identify mRNA-CLU levels and CLU immunostaining in the spermatozoa acrosome as independent risk factors for pregnancy failure, with distribution patterns correlating with sperm maturity and seminogram alterations.


Sujet(s)
Clusterine , Spermatozoïdes , Humains , Clusterine/métabolisme , Clusterine/génétique , Mâle , Spermatozoïdes/métabolisme , Adulte , Femelle , Fécondité/physiologie , Grossesse , Fécondation in vitro , Infertilité masculine/métabolisme , ARN messager/métabolisme , ARN messager/génétique
10.
N Engl J Med ; 390(23): 2217-2219, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38899702
11.
Neurosci Lett ; 836: 137874, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-38857696

RÉSUMÉ

Clusterin is a secreted glycoprotein that participates in multiple physiological processes through its chaperon function. In Alzheimer's disease, the brain functions under an increased oxidative stress condition that causes an elevation of protein oxidation, resulting in enhanced pathology. Accordingly, it is important to determine the type of human brain cells that are mostly prone to methionine oxidation in Alzheimer's disease and specifically monitoring the methionine-oxidation levels of clusterin in human and mice brains and its effect on clusterin's function. We analyzed the level of methionine sulfoxide (MetO)-clusterin in these brains, using a combination of immunoprecipitation and Western-blott analyses. Also, we determine the effect of methionine oxidation on clusterin ability to bind beta-amyloid, in vitro, using calorimetric assay. Our results show that human neurons and astrocytes of Alzheimer's disease brains are mostly affected by methionine oxidation. Moreover, MetO-clusterin levels are elevated in postmortem Alzheimer's disease human and mouse brains in comparison to controls. Finally, oxidation of methionine residues of purified clusterin reduced its binding efficiency to beta-amyloid. In conclusion, we suggest that methionine oxidation of brain-clusterin is enhanced in Alzheimer's disease and that this oxidation compromises its chaperon function, leading to exacerbation of beta-amyloid's toxicity in Alzheimer's disease.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Astrocytes , Encéphale , Clusterine , Méthionine , Oxydoréduction , Sujet âgé , Animaux , Humains , Mâle , Souris , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Astrocytes/métabolisme , Encéphale/métabolisme , Clusterine/métabolisme , Méthionine/métabolisme , Méthionine/analogues et dérivés , Neurones/métabolisme , Liaison aux protéines
12.
Int Immunopharmacol ; 137: 112355, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38851158

RÉSUMÉ

One major obstacle in the treatment of cancer is the presence of proteins resistant to cancer therapy, which can impede the effectiveness of traditional approaches such as radiation and chemotherapy. This resistance can lead to disease progression and cause treatment failure. Extensive research is currently focused on studying these proteins to create tailored treatments that can circumvent resistance mechanisms. CLU (Clusterin), a chaperone protein, has gained notoriety for its role in promoting resistance to a wide range of cancer treatments, including chemotherapy, radiation therapy, and targeted therapy. The protein has also been discovered to have a role in regulating the immunosuppressive environment within tumors. Its ability to influence oncogenic signaling and inhibit cell death bolster cancer cells resistant against treatments, which poses a significant challenge in the field of oncology. Researchers are actively investigating to the mechanisms by which CLU exerts its resistance-promoting effects, with the ultimate goal of developing strategies to circumvent its impact and enhance the effectiveness of cancer therapies. By exploring CLU's impact on cancer, resistance mechanisms, tumor microenvironment (TME), and therapeutic strategies, this review aims to contribute to the ongoing efforts to improve cancer treatment outcomes.


Sujet(s)
Clusterine , Résistance aux médicaments antinéoplasiques , Tumeurs , Microenvironnement tumoral , Humains , Clusterine/métabolisme , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Animaux , Microenvironnement tumoral/immunologie
13.
J Alzheimers Dis ; 99(4): 1261-1271, 2024.
Article de Anglais | MEDLINE | ID: mdl-38788070

RÉSUMÉ

Background: Reliable blood biomarkers are crucial for early detection and treatment evaluation of cognitive impairment, including Alzheimer's disease and other dementias. Objective: To examine whether plasma biomarkers and their combination are different between older people with mild cognitive impairment (MCI) and cognitively normal individuals, and to explore their relations with cognitive performance. Methods: This cross-sectional study included 250 older adults, including 124 participants with MCI, and 126 cognitively normal participants. Plasma brain-derived neurotrophic factor (BDNF), irisin and clusterin were measured, and BDNF/irisin ratio was calculated. Global cognition was evaluated by the Montreal Cognitive Assessment. Results: Plasma irisin levels, but not BDNF, were significantly different between MCI group and cognitively normal group. Higher irisin concentration was associated with an increased probability for MCI both before and after controlling covariates. By contrast, plasma BDNF concentration, but not irisin, was linearly correlated with cognitive performance after adjusting for covariates. Higher BDNF/irisin ratios were not only correlated with better cognitive performance, but also associated with lower risks of MCI, no matter whether we adjusted for covariates. Plasma BDNF and irisin concentrations increased with aging, whereas BDNF/irisin ratios remained stable. No significant results of clusterin were observed. Conclusions: Plasma BDNF/irisin ratio may be a reliable indicator which not only reflects the odds of the presence of MCI but also directly associates with cognitive performance.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Clusterine , Cognition , Dysfonctionnement cognitif , Fibronectines , Humains , Facteur neurotrophique dérivé du cerveau/sang , Mâle , Femelle , Sujet âgé , Dysfonctionnement cognitif/sang , Dysfonctionnement cognitif/diagnostic , Fibronectines/sang , Études transversales , Clusterine/sang , Cognition/physiologie , Sujet âgé de 80 ans ou plus , Marqueurs biologiques/sang , Tests neuropsychologiques/statistiques et données numériques , Vieillissement/sang , Tests de l'état mental et de la démence
15.
Cells ; 13(8)2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38667280

RÉSUMÉ

Clusterin (CLU) protein is involved in various pathophysiological processes including carcinogenesis and tumor progression. In recent years, the role of the secretory isoform has been demonstrated in tumor cells, where it inhibits apoptosis and favors the acquisition of resistance to conventional treatments used to treat cancer. To determine the possible therapeutic potential of inhibiting this protein, numerous studies have been carried out in this field. In this article, we present the existing knowledge to date on the inhibition of this protein in different types of cancer and analyze the importance it could have in the development of new therapies targeted against this disease.


Sujet(s)
Clusterine , Tumeurs , Clusterine/métabolisme , Clusterine/antagonistes et inhibiteurs , Humains , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie
16.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38673784

RÉSUMÉ

COVID-19 is an infectious disease caused by the SARS-CoV-2 virus. Glycoprotein clusterin (CLU) has many functions such as phagocyte recruitment, complement system inhibition, apoptosis inhibition, hormone and lipid transport, as well as in the immune response. The study aimed to assess the changes in CLU concentrations and the profile and degree of CLU glycosylation between patients with severe COVID-19, convalescents, and healthy subjects (control). The profile and degree of serum CLU N-glycosylation were analyzed using lectin-ELISA with specific lectins. CLU concentrations were significantly lower and relative reactivities of CLU glycans with SNA (Sambucus nigra agglutinin) were significantly higher in severe COVID-19 patients in comparison to convalescents and the control group. The relative reactivities of CLU glycans with MAA (Maackia amurensis agglutinin), together with relative reactivity with LCA (Lens culinaris agglutinin), were also significantly higher in patients with severe COVID-19 than in convalescents and the control group, but they also significantly differed between convalescents and control. The development of acute inflammation in the course of severe COVID-19 is associated with a decrease in CLU concentration, accompanied by an increase in the expression of α2,3-linked sialic acid, and core fucose. Both of these parameters can be included as useful glycomarkers differentiating patients with severe COVID-19 from convalescents and the control group, as well as convalescents and healthy subjects.


Sujet(s)
Marqueurs biologiques , COVID-19 , Clusterine , SARS-CoV-2 , Femelle , Humains , Mâle , Marqueurs biologiques/sang , Clusterine/sang , COVID-19/sang , COVID-19/diagnostic , Glycosylation , Lectines/sang
17.
Int J Biol Macromol ; 266(Pt 2): 131341, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38574922

RÉSUMÉ

Sialic acids are negatively charged carbohydrates that are components of saccharide chains covalently linked to macromolecules. Sialylated glycoproteins are important for most biological processes, including reproduction, where they are associated with spermatogenesis, sperm motility, immune responses, and fertilization. Changes in the glycoprotein profile or sialylation in glycoproteins are likely to affect the quality of ejaculate. The aim of this study was to determine differences in the degree of sialylation between normozoospermic ejaculates and ejaculates with a pathological spermiogram using two lectins, Sambucus nigra (SNA) and Maackia amurensis (MAL II/MAA) recognizing α-2,6 or α-2,3 linkage of Sia to galactosyl residues. Our results show a close relationship between seminal plasma (SP) sialoproteins and the presence of anti-sperm antibodies in the ejaculate, apoptotic spermatozoa, and ejaculate quality. Using mass spectrometry, we identified SP sialoproteins such as, semenogelins, glycodelin, prolactin-inducible protein, lactotransferrin, and clusterin that are associated with spermatozoa and contribute to the modulation of the immune response and sperm apoptosis. Our findings suggest a correlation between the degree of SP glycoprotein sialylation and the existence of possible pathological states of spermatozoa and reproductive organs. Glycoproteins sialylation represents a potential parameter reflecting the overall quality of ejaculate and could potentially be utilised in diagnostics.


Sujet(s)
Sperme , Spermatozoïdes , Mâle , Humains , Sperme/métabolisme , Sperme/composition chimique , Spermatozoïdes/métabolisme , Mobilité des spermatozoïdes , Glycoprotéines/métabolisme , Glycodeline/métabolisme , Protéines sécrétoires des vésicules séminales/métabolisme , Analyse du sperme/méthodes , Clusterine/métabolisme , Lectines/métabolisme , Lectines/composition chimique , Éjaculation , Acides sialiques/métabolisme , Protéines du plasma séminal/métabolisme , Lactoferrine/métabolisme , Apoptose
18.
Autophagy ; 20(6): 1359-1382, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38447939

RÉSUMÉ

Mitophagy involves the selective elimination of defective mitochondria during chemotherapeutic stress to maintain mitochondrial homeostasis and sustain cancer growth. Here, we showed that CLU (clusterin) is localized to mitochondria to induce mitophagy controlling mitochondrial damage in oral cancer cells. Moreover, overexpression and knockdown of CLU establish its mitophagy-specific role, where CLU acts as an adaptor protein that coordinately interacts with BAX and LC3 recruiting autophagic machinery around damaged mitochondria in response to cisplatin treatment. Interestingly, CLU triggers class III phosphatidylinositol 3-kinase (PtdIns3K) activity around damaged mitochondria, and inhibition of mitophagic flux causes the accumulation of excessive mitophagosomes resulting in reactive oxygen species (ROS)-dependent apoptosis during cisplatin treatment in oral cancer cells. In parallel, we determined that PPARGC1A/PGC1α (PPARG coactivator 1 alpha) activates mitochondrial biogenesis during CLU-induced mitophagy to maintain the mitochondrial pool. Intriguingly, PPARGC1A inhibition through small interfering RNA (siPPARGC1A) and pharmacological inhibitor (SR-18292) treatment counteracts CLU-dependent cytoprotection leading to mitophagy-associated cell death. Furthermore, co-treatment of SR-18292 with cisplatin synergistically suppresses tumor growth in oral cancer xenograft models. In conclusion, CLU and PPARGC1A are essential for sustained cancer cell growth by activating mitophagy and mitochondrial biogenesis, respectively, and their inhibition could provide better therapeutic benefits against oral cancer.


Sujet(s)
Survie cellulaire , Clusterine , Mitochondries , Mitophagie , Tumeurs de la bouche , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Humains , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Clusterine/métabolisme , Clusterine/génétique , Mitophagie/effets des médicaments et des substances chimiques , Mitophagie/physiologie , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/métabolisme , Tumeurs de la bouche/génétique , Animaux , Survie cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Cisplatine/pharmacologie , Biogenèse des organelles , Souris , Apoptose/effets des médicaments et des substances chimiques , Souris nude , Espèces réactives de l'oxygène/métabolisme , Autophagie/physiologie , Autophagie/effets des médicaments et des substances chimiques
19.
PLoS One ; 19(2): e0298133, 2024.
Article de Anglais | MEDLINE | ID: mdl-38363768

RÉSUMÉ

Alterations in von Willebrand factor (VWF) have an important role in human health and disease. Deficiency of VWF is associated with symptoms of bleeding and excesses of VWF are associated with thrombotic outcomes. Understanding the mechanisms that drive VWF regulation can lead to a better understanding of modulation of VWF levels in humans. We identified clusterin (CLU) as a potential candidate regulator of VWF based on a single cell RNA sequencing (scRNA-seq) analysis in control endothelial cells (ECs) and von Willebrand disease (VWD) endothelial colony-forming-cells (ECFCs). We found that patients with deficiencies of VWF (von Willebrand disease, VWD) had decreased CLU expression and ECs with low VWF expression also had low CLU expression. Based on these findings, we sought to evaluate the role of CLU in the regulation of VWF, specifically as it relates to VWD. As CLU is primarily thought to be a golgi protein involved in protein chaperoning, we hypothesized that knockdown of CLU would lead to decreases in VWF and alterations in Weibel-Palade bodies (WPBs). We used both siRNA- and CRISPR-Cas9-based approaches to modulate CLU in human umbilical vein endothelial cells (HUVECs) and evaluated VWF protein levels, VWF mRNA copy number, and WPB quantity and size. We demonstrated that siRNA-based knockdown of CLU resulted in decreases in VWF content in cellular lysates and supernatants, but no significant change in WPB quantity or size. A CRISPR-Cas9-based knockdown of CLU demonstrated similar findings of decreases in intracellular VWF content but no significant change in WPB quantity or size. Our data suggests that CLU knockdown is associated with decreases in cellular VWF content but does not affect VWF mRNA levels or WPB quantity or size.


Sujet(s)
Clusterine , Facteur de von Willebrand , Humains , Cellules cultivées , Clusterine/génétique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Maladies de von Willebrand , Facteur de von Willebrand/génétique , Facteur de von Willebrand/métabolisme , Corps de Weibel-Palade/métabolisme
20.
Cancer Metastasis Rev ; 43(1): 379-391, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38319453

RÉSUMÉ

Intra-tumoural heterogeneity and cancer cell plasticity in colorectal cancer (CRC) have been key challenges to effective treatment for patients. It has been suggested that a subpopulation of LGR5-expressing cancer stem cells (CSCs) is responsible for driving tumour relapse and therapy resistance in CRC. However, studies have revealed that the LGR5+ve CSC population is highly sensitive to chemotherapy. It has been hypothesised that another subset of tumour cells can phenotypically revert to a stem-like state in response to chemotherapy treatment which replenishes the LGR5+ve CSC population and maintains tumour growth. Recently, a unique stem cell population marked by enriched clusterin (CLU) expression and termed the revival stem cell (RevSC) was identified in the regenerating murine intestine. This CLU-expressing cell population is quiescent during homeostasis but has the ability to survive and regenerate other stem cells upon injury. More recently, the CLU+ve signature has been implicated in several adverse outcomes in CRC, including chemotherapy resistance and poor patient survival; however, the mechanism behind this remains undetermined. In this review, we discuss recent insights on CLU in CRC and its roles in enhancing the plasticity of cells and further consider the implications of CLU as a prospective target for therapeutic intervention.


Sujet(s)
Tumeurs colorectales , Résistance aux médicaments antinéoplasiques , Animaux , Humains , Clusterine/métabolisme , Tumeurs colorectales/anatomopathologie , Cellules souches tumorales/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE