Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.162
Filter
1.
Cells ; 13(17)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39273003

ABSTRACT

TRIM44, a tripartite motif (TRIM) family member, is pivotal in linking the ubiquitin-proteasome system (UPS) to autophagy in multiple myeloma (MM). However, its prognostic impact and therapeutic potential remain underexplored. Here, we report that TRIM44 overexpression is associated with poor prognosis in a Multiple Myeloma Research Foundation (MMRF) cohort of 858 patients, persisting across primary and recurrent MM cases. TRIM44 expression notably increases in advanced MM stages, indicating its potential role in disease progression. Single-cell RNA sequencing across MM stages showed significant TRIM44 upregulation in smoldering MM (SMM) and MM compared to normal bone marrow, especially in patients with t(4;14) cytogenetic abnormalities. This analysis further identified high TRIM44 expression as predictive of lower responsiveness to proteasome inhibitor (PI) treatments, underscoring its critical function in the unfolded protein response (UPR) in TRIM44-high MM cells. Our findings also demonstrate that TRIM44 facilitates SQSTM1 oligomerization under oxidative stress, essential for its phosphorylation and subsequent autophagic degradation. This process supports the survival of PI-resistant MM cells by activating the NRF2 pathway, which is crucial for oxidative stress response and, potentially, other chemotherapy-induced stressors. Additionally, TRIM44 counters the TRIM21-mediated suppression of the antioxidant response, enhancing MM cell survival under oxidative stress. Collectively, our discoveries highlight TRIM44's significant role in MM progression and resistance to therapy, suggesting its potential value as a therapeutic target.


Subject(s)
Multiple Myeloma , Proteasome Endopeptidase Complex , Tripartite Motif Proteins , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/genetics , Humans , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Prognosis , Cell Line, Tumor , Proteasome Endopeptidase Complex/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Autophagy/genetics , Cell Survival/drug effects , Cell Survival/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Proteasome Inhibitors/pharmacology , Drug Resistance, Neoplasm/genetics , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Gene Expression Regulation, Neoplastic
2.
Stem Cell Res ; 80: 103520, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39126919

ABSTRACT

SQSTM1 (Sequestosome 1) also known as p62, plays several important physiological roles in the cell. It regulates autophagy and mitochondrial homeostasis and can further lead to metabolic reprogramming. Pathogenic variants in SQSTM1 gene are known to cause Neurodegeneration with ataxia, dystonia, and gaze palsy in autosomal recessive inheritance fashion. We report here, the generation of induced pluripotent stem cell (iPSC) line (IGIBi010-A) carrying a novel homozygous frameshift variant in SQSTM1 i.e. p.Leu251SerfsTer4. In future, this iPSC line will be used as a resource to elucidate the molecular pathway, targeting strategies for disease biology derived by variation in SQSTM1 gene.


Subject(s)
Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Phenotype , Sequestosome-1 Protein , Humans , Induced Pluripotent Stem Cells/metabolism , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Cell Line , Mutation , Male , Female
3.
Redox Rep ; 29(1): 2392329, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39150892

ABSTRACT

Doxorubicin (Dox) is extensively used as an antitumor agent, but its severe cardiotoxicity significantly limits its clinical use. Current treatments for Dox-induced cardiotoxicity are inadequate, necessitating alternative solutions. This study evaluated the effects of sarmentosin, a compound from Sedum sarmentosum, on Dox-induced cardiotoxicity and dysfunction. Sarmentosin was administered as a pretreatment to both mice and H9c2 cells before Dox exposure. Subsequently, markers of Dox-induced cardiotoxicity and ferroptosis in serum and cell supernatants were measured. Western blot analysis was utilized to detect levels of ferroptosis, oxidative stress, and autophagy proteins. Additionally, echocardiography, hematoxylin-eosin staining, ROS detection, and immunofluorescence techniques were employed to support our findings. Results demonstrated that sarmentosin significantly inhibited iron accumulation, lipid peroxidation, and oxidative stress, thereby reducing Dox-induced ferroptosis and cardiotoxicity in C57BL/6 mice and H9c2 cells. The mechanism involved the activation of autophagy and the Nrf2 signaling pathway. These findings suggest that sarmentosin may prevent Dox-induced cardiotoxicity by mitigating ferroptosis. The study underscores the potential of compounds like sarmentosin in treating Dox-induced cardiotoxicity.


Subject(s)
Cardiotoxicity , Doxorubicin , Ferroptosis , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Signal Transduction , Animals , Ferroptosis/drug effects , NF-E2-Related Factor 2/metabolism , Doxorubicin/adverse effects , Doxorubicin/toxicity , Mice , Cardiotoxicity/drug therapy , Cardiotoxicity/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Signal Transduction/drug effects , Oxidative Stress/drug effects , Male , Mice, Inbred C57BL , Autophagy/drug effects , Sequestosome-1 Protein/metabolism
4.
Sci Rep ; 14(1): 18974, 2024 08 16.
Article in English | MEDLINE | ID: mdl-39152142

ABSTRACT

The deubiquitinase tripartite motif containing 44 (TRIM44) plays a critical role in linking the proteotoxic stress response with autophagic degradation, which is significant in the context of cancer and neurological diseases. Although TRIM44 is recognized as a prognostic marker in various cancers, the complex molecular mechanisms through which it facilitates autophagic degradation, particularly under oxidative stress conditions, have not been fully explored. In this study, we demonstrate that TRIM44 significantly enhances autophagy in response to oxidative stress, reducing cytotoxicity in cancer cells treated with arsenic trioxide. Our research emphasizes the critical role of the posttranslational modification of sequestosome-1 (SQSTM1) and its importance in improving sequestration during autophagic degradation under oxidative stress. We found that TRIM44 notably promotes SQSTM1 oligomerization in both PB1 domain-dependent and oxidation-dependent manners. Furthermore, TRIM44 amplifies the interaction between protein kinase A and oligomerized SQSTM1, leading to enhanced phosphorylation of SQSTM1 at S349. This phosphorylation event activates NFE2L2, a key transcription factor in the oxidative stress response, highlighting the importance of TRIM44 in modulating SQSTM1-mediated autophagy. Our findings support that TRIM44 plays pivotal roles in regulating autophagic sensitivity to oxidative stress, with implications for cancer, aging, aging-associated diseases, and neurodegenerative disorders.


Subject(s)
Autophagy , NF-E2-Related Factor 2 , Oxidative Stress , Sequestosome-1 Protein , Tripartite Motif Proteins , Sequestosome-1 Protein/metabolism , Humans , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , NF-E2-Related Factor 2/metabolism , Phosphorylation , Arsenic Trioxide/pharmacology , Protein Multimerization , Cell Line, Tumor , Intracellular Signaling Peptides and Proteins/metabolism , HEK293 Cells
5.
Cells ; 13(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39120287

ABSTRACT

Autophagy engulfs cellular components in double-membrane-bound autophagosomes for clearance and recycling after fusion with lysosomes. Thus, autophagy is a key process for maintaining proteostasis and a powerful cell-intrinsic host defense mechanism, protecting cells against pathogens by targeting them through a specific form of selective autophagy known as xenophagy. In this context, ubiquitination acts as a signal of recognition of the cargoes for autophagic receptors, which direct them towards autophagosomes for subsequent breakdown. Nevertheless, autophagy can carry out a dual role since numerous viruses including members of the Orthoherpesviridae family can either inhibit or exploit autophagy for its own benefit and to replicate within host cells. There is growing evidence that Herpes simplex virus type 1 (HSV-1), a highly prevalent human pathogen that infects epidermal keratinocytes and sensitive neurons, is capable of negatively modulating autophagy. Since the effects of HSV-1 infection on autophagic receptors have been poorly explored, this study aims to understand the consequences of HSV-1 productive infection on the levels of the major autophagic receptors involved in xenophagy, key proteins in the recruitment of intracellular pathogens into autophagosomes. We found that productive HSV-1 infection in human neuroglioma cells and keratinocytes causes a reduction in the total levels of Ub conjugates and decreases protein levels of autophagic receptors, including SQSTM1/p62, OPTN1, NBR1, and NDP52, a phenotype that is also accompanied by reduced levels of LC3-I and LC3-II, which interact directly with autophagic receptors. Mechanistically, we show these phenotypes are the result of xenophagy activation in the early stages of productive HSV-1 infection to limit virus replication, thereby reducing progeny HSV-1 yield. Additionally, we found that the removal of the tegument HSV-1 protein US11, a recognized viral factor that counteracts autophagy in host cells, enhances the clearance of autophagic receptors, with a significant reduction in the progeny HSV-1 yield. Moreover, the removal of US11 increases the ubiquitination of SQSTM1/p62, indicating that US11 slows down the autophagy turnover of autophagy receptors. Overall, our findings suggest that xenophagy is a potent host defense against HSV-1 replication and reveals the role of the autophagic receptors in the delivery of HSV-1 to clearance via xenophagy.


Subject(s)
Autophagy , Herpesvirus 1, Human , Humans , Herpesvirus 1, Human/physiology , Herpes Simplex/virology , Herpes Simplex/immunology , Herpes Simplex/metabolism , Macroautophagy , Virus Replication , Autophagosomes/metabolism , Keratinocytes/virology , Keratinocytes/metabolism , Sequestosome-1 Protein/metabolism , Host-Pathogen Interactions , Animals , Nuclear Proteins , Cell Cycle Proteins , Membrane Transport Proteins
6.
Int J Mol Sci ; 25(15)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39125836

ABSTRACT

Regulation of autophagy through the 62 kDa ubiquitin-binding protein/autophagosome cargo protein sequestosome 1 (p62/SQSTM1), whose level is generally inversely proportional to autophagy, is crucial in microglial functions. Since autophagy is involved in inflammatory mechanisms, we investigated the actions of pro-inflammatory lipopolysaccharide (LPS) and anti-inflammatory rosuvastatin (RST) in secondary microglial cultures with or without bafilomycin A1 (BAF) pretreatment, an antibiotic that potently inhibits autophagosome fusion with lysosomes. The levels of the microglia marker protein Iba1 and the autophagosome marker protein p62/SQSTM1 were quantified by Western blots, while the number of p62/SQSTM1 immunoreactive puncta was quantitatively analyzed using fluorescent immunocytochemistry. BAF pretreatment hampered microglial survival and decreased Iba1 protein level under all culturing conditions. Cytoplasmic p62/SQSTM1 level was increased in cultures treated with LPS+RST but reversed markedly when BAF+LPS+RST were applied together. Furthermore, the number of p62/SQSTM1 immunoreactive autophagosome puncta was significantly reduced when RST was used but increased significantly in BAF+RST-treated cultures, indicating a modulation of autophagic flux through reduction in p62/SQSTM1 degradation. These findings collectively indicate that the cytoplasmic level of p62/SQSTM1 protein and autophagocytotic flux are differentially regulated, regardless of pro- or anti-inflammatory state, and provide context for understanding the role of autophagy in microglial function in various inflammatory settings.


Subject(s)
Autophagosomes , Autophagy , Lipopolysaccharides , Macrolides , Microglia , Sequestosome-1 Protein , Animals , Sequestosome-1 Protein/metabolism , Microglia/metabolism , Microglia/drug effects , Macrolides/pharmacology , Autophagy/drug effects , Rats , Autophagosomes/metabolism , Autophagosomes/drug effects , Lipopolysaccharides/pharmacology , Cells, Cultured , Inflammation/metabolism , Biomarkers/metabolism
7.
J Nanobiotechnology ; 22(1): 464, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095755

ABSTRACT

BACKGROUND: Doxorubicin (DOX) is a first-line chemotherapeutic drug for various malignancies that causes cardiotoxicity. Plant-derived exosome-like nanovesicles (P-ELNs) are growing as novel therapeutic agents. Here, we investigated the protective effects in DOX cardiotoxicity of ELNs from Momordica charantia L. (MC-ELNs), a medicinal plant with antioxidant activity. RESULTS: We isolated MC-ELNs using ultracentrifugation and characterized them with canonical mammalian extracellular vesicles features. In vivo studies proved that MC-ELNs ameliorated DOX cardiotoxicity with enhanced cardiac function and myocardial structure. In vitro assays revealed that MC-ELNs promoted cell survival, diminished reactive oxygen species, and protected mitochondrial integrity in DOX-treated H9c2 cells. We found that DOX treatment decreased the protein level of p62 through ubiquitin-dependent degradation pathway in H9c2 and NRVM cells. However, MC-ELNs suppressed DOX-induced p62 ubiquitination degradation, and the recovered p62 bound with Keap1 promoting Nrf2 nuclear translocation and the expressions of downstream gene HO-1. Furthermore, both the knockdown of Nrf2 and the inhibition of p62-Keap1 interaction abrogated the cardioprotective effect of MC-ELNs. CONCLUSIONS: Our findings demonstrated the therapeutic beneficials of MC-ELNs via increasing p62 protein stability, shedding light on preventive approaches for DOX cardiotoxicity.


Subject(s)
Cardiotoxicity , Doxorubicin , Exosomes , Momordica charantia , NF-E2-Related Factor 2 , Animals , Cardiotoxicity/prevention & control , Cardiotoxicity/metabolism , Momordica charantia/chemistry , Exosomes/metabolism , Rats , NF-E2-Related Factor 2/metabolism , Cell Line , Kelch-Like ECH-Associated Protein 1/metabolism , Reactive Oxygen Species/metabolism , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Cell Survival/drug effects , Rats, Sprague-Dawley , Sequestosome-1 Protein/metabolism
8.
Int Immunopharmacol ; 141: 112934, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39178516

ABSTRACT

Hepatic fibrosis is a common pathology present in most chronic liver diseases. Autophagy is a lysosome-mediated intracellular catabolic and recycling process that plays an essential role in maintaining normal hepatic functions. Nuclear factor erythroid 2-like 2 (Nrf2) is a transcription factor responsible for the regulation of cellular anti-oxidative stress response. This study was designed to assess the cytoprotective effect of mesenchymal stem cell-derived exosomes (MSC-exos) on endothelial-mesenchymal transition (EMT) in Carbon Tetrachloride (CCL4) induced liver fibrosis. Rats were treated with 0.1 ml of CCL4 twice weekly for 8 weeks, followed by administration of a single dose of MSC-exos. Rats were then sacrificed after 4 weeks, and liver samples were collected for gene expression analyses, Western blot, histological studies, immunohistochemistry, and transmission electron microscopy. Our results showed that MSC-exos administration decreased collagen deposition, apoptosis, and inflammation. Exosomes modulate the Nrf2/Keap1/p62 pathway, restoring autophagy and Nrf2 levels through modulation of the non-canonical pathway of Nrf2/Keap1/p62. Additionally, MSC-exos regulated miR-153-3p, miR-27a, miR-144 and miRNA-34a expression. In conclusion, the present study shed light on MSC-exos as a cytoprotective agent against EMT and tumorigenesis in chronic liver inflammation.


Subject(s)
Carbon Tetrachloride , Exosomes , Kelch-Like ECH-Associated Protein 1 , Liver Cirrhosis , Mesenchymal Stem Cells , MicroRNAs , NF-E2-Related Factor 2 , Signal Transduction , Animals , Exosomes/metabolism , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Mesenchymal Stem Cells/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/therapy , Male , Rats , MicroRNAs/metabolism , MicroRNAs/genetics , Rats, Sprague-Dawley , Liver/pathology , Liver/metabolism , Autophagy , Sequestosome-1 Protein/metabolism
9.
Mol Cell ; 84(17): 3271-3287.e8, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39178863

ABSTRACT

Cellular senescence, a stress-induced stable proliferation arrest associated with an inflammatory senescence-associated secretory phenotype (SASP), is a cause of aging. In senescent cells, cytoplasmic chromatin fragments (CCFs) activate SASP via the anti-viral cGAS/STING pathway. Promyelocytic leukemia (PML) protein organizes PML nuclear bodies (NBs), which are also involved in senescence and anti-viral immunity. The HIRA histone H3.3 chaperone localizes to PML NBs in senescent cells. Here, we show that HIRA and PML are essential for SASP expression, tightly linked to HIRA's localization to PML NBs. Inactivation of HIRA does not directly block expression of nuclear factor κB (NF-κB) target genes. Instead, an H3.3-independent HIRA function activates SASP through a CCF-cGAS-STING-TBK1-NF-κB pathway. HIRA physically interacts with p62/SQSTM1, an autophagy regulator and negative SASP regulator. HIRA and p62 co-localize in PML NBs, linked to their antagonistic regulation of SASP, with PML NBs controlling their spatial configuration. These results outline a role for HIRA and PML in the regulation of SASP.


Subject(s)
Cell Cycle Proteins , Cellular Senescence , Histone Chaperones , Inflammation , NF-kappa B , Nuclear Proteins , Promyelocytic Leukemia Protein , Protein Serine-Threonine Kinases , Sequestosome-1 Protein , Signal Transduction , Transcription Factors , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Autophagy , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Chromatin/metabolism , Chromatin/genetics , HEK293 Cells , Histone Chaperones/metabolism , Histone Chaperones/genetics , Histones/metabolism , Histones/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , NF-kappa B/metabolism , NF-kappa B/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nucleotidyltransferases , Promyelocytic Leukemia Protein/metabolism , Promyelocytic Leukemia Protein/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics
10.
Life Sci ; 356: 122981, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39147314

ABSTRACT

Doxorubicin-induced cardiotoxicity (DIC) poses a significant challenge, impeding its widespread application. Emerging evidence suggests the involvement of ferroptosis in the DIC. While the downregulation of SLC7A11 expression has been linked to the promotion of ferroptosis, the precise regulatory mechanism remains unclear. Recent studies, including our own, have highlighted abnormal levels of autophagy adapter protein P62 and autophagy in DIC development. Thus, our study aimed to further investigate the role of autophagy and ferroptosis in DIC, elucidating underlying molecular mechanisms across molecular, cellular, and whole-organ levels utilizing gene knockdown, immunoprecipitation, and mass spectrometry techniques. The results of our findings unveiled cardiomyocyte damage, heightened autophagy levels, and ferroptosis in DOX-treated mouse hearts. Notably, inhibition of autophagy levels attenuated DOX-induced ferroptosis. Mechanistically, we discovered that the autophagy adaptor protein P62 mediates the entry of SLC7A11 into the autophagic pathway for degradation. Furthermore, the addition of autophagy inhibitors (CQ or BAF) could elevate SLC7A11 and GPX4 protein expression, reduce the accumulation of Fe2+ and ROS in cardiomyocytes, and thus mitigate DOX-induced ferroptosis. In summary, our findings underscore the pivotal role of the P62-autophagy pathway in SLC7A11 degradation, modulating ferroptosis to exacerbate DIC. This finding offers significant insights into the underlying molecular mechanisms of DOX-induced ferroptosis and identifies new targets for reversing DIC.


Subject(s)
Amino Acid Transport System y+ , Autophagy , Cardiotoxicity , Doxorubicin , Ferroptosis , Myocytes, Cardiac , Sequestosome-1 Protein , Animals , Male , Mice , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Antibiotics, Antineoplastic/toxicity , Antibiotics, Antineoplastic/adverse effects , Autophagy/drug effects , Cardiotoxicity/metabolism , Cardiotoxicity/etiology , Doxorubicin/adverse effects , Doxorubicin/toxicity , Ferroptosis/drug effects , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics
11.
Methods Mol Biol ; 2845: 191-196, 2024.
Article in English | MEDLINE | ID: mdl-39115667

ABSTRACT

p62 bodies are ubiquitin-positive cytoplasmic condensates formed by liquid-liquid phase separation. They are targeted by selective autophagy and play important roles in intracellular quality control and stress responses. However, little is known about their constituents. In this chapter, we describe a method for purifying p62 bodies using fluorescence-activated particle sorting. This method contributes to the identification of novel components of p62 bodies under various physiological and stress conditions.


Subject(s)
Autophagy , Flow Cytometry , Humans , Flow Cytometry/methods , Ubiquitin/metabolism , Sequestosome-1 Protein/metabolism
12.
Anal Cell Pathol (Amst) ; 2024: 5573892, 2024.
Article in English | MEDLINE | ID: mdl-39131899

ABSTRACT

Among autophagic-related proteins, p62/SQSTM1/Sequestosome-1 represents a relevant actor in cellular proliferation and neoplastic growth. Although, recently, p62 expression has been analyzed in different neurodegenerative and glial neoplastic diseases, no available information have been reported in meningiomas, which have an high epidemiological relevance being the second most common category of intracranial tumors after gliomas. Generally meningiomas have a benign behavior, but their recurrence is not uncommon mainly when atypical or anaplastic varieties occur. However, intranuclear vacuoles have been ultrastructurally observed in meningiomas, and they were labelled by p62 antibodies. Therefore, in the present study, we have investigated p62 immunohistochemical pattern in a cohort of 133 cases representative of low- and high-grade meningiomas, to verify if p62 expression may be related to clinicopathological data, thus achieving a potential prognostic role. The p62 immunoexpression was frequently found in the nucleus and cytoplasm of neoplastic elements, and utilizing an intensity-distribution score, 55 (41.3%) cases were considered as high expressors while 78 (58.7%) cases were instead recorded as low expressors. Fifteen cases exhibited recurrences of the disease, 14 of which were codified as high expressors. Moreover, a direct relationship between p62 and Mib-1 immunoexpression as well as between p62 and neoplastic grade have been documented. Finally, we suggest that impaired autophagic flux with an increase in p62 expression may be involved in the activation of NRF2 also contributing in the development of recurrence in meningioma patients.


Subject(s)
Immunohistochemistry , Meningeal Neoplasms , Meningioma , Neoplasm Grading , Sequestosome-1 Protein , Humans , Meningioma/metabolism , Meningioma/pathology , Sequestosome-1 Protein/metabolism , Female , Male , Middle Aged , Aged , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Adult , Aged, 80 and over , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology
13.
Vascul Pharmacol ; 156: 107417, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39159737

ABSTRACT

Myocardial infarction (MI) and the ensuing heart failure (HF) remain the main cause of morbidity and mortality worldwide. One of the strategies to combat MI and HF lies in the ability to accurately predict the onset of these disorders. Alterations in mitochondrial homeostasis have been reported to be involved in the pathogenesis of various cardiovascular diseases (CVDs). In this regard, perturbations to mitochondrial dynamics leading to impaired clearance of dysfunctional mitochondria have been previously established to be a crucial trigger for MI/HF. In this study, we found that MI patients could be classified into three clusters based on the expression levels of mitophagy-related genes and consensus clustering. We identified a mitophagy-related diagnostic 5-genes signature for MI using support vector machines-Recursive Feature Elimination (SVM-RFE) and random forest, with the area under the ROC curve (AUC) value of the predictive model at 0.813. Additionally, the single-cell transcriptome and pseudo-time analyses showed that the mitoscore was significantly upregulated in macrophages, endothelial cells, pericytes, fibroblasts and monocytes in patients with ischemic cardiomyopathy, while sequestosome 1 (SQSTM1) exhibited remarkable increase in the infarcted (ICM) and non-infarcted (ICMN) myocardium samples dissected from the left ventricle compared with control samples. Lastly, through analysis of peripheral blood from MI patients, we found that the expression of SQSTM1 is positively correlated with troponin-T (P < 0.0001, R = 0.4195, R2 = 0.1759). Therefore, this study provides the rationale for a cell-specific mitophagy-related gene signature as an additional supporting diagnostic for CVDs.


Subject(s)
Gene Expression Profiling , Mitophagy , Myocardial Infarction , Predictive Value of Tests , Transcriptome , Mitophagy/genetics , Humans , Myocardial Infarction/genetics , Myocardial Infarction/diagnosis , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Male , Middle Aged , Female , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondria, Heart/genetics , Aged , Support Vector Machine , Genetic Markers , Case-Control Studies
14.
Science ; 385(6712): eadj7446, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39208097

ABSTRACT

Chromosomal instability (CIN) generates micronuclei-aberrant extranuclear structures that catalyze the acquisition of complex chromosomal rearrangements present in cancer. Micronuclei are characterized by persistent DNA damage and catastrophic nuclear envelope collapse, which exposes DNA to the cytoplasm. We found that the autophagic receptor p62/SQSTM1 modulates micronuclear stability, influencing chromosome fragmentation and rearrangements. Mechanistically, proximity of micronuclei to mitochondria led to oxidation-driven homo-oligomerization of p62, limiting endosomal sorting complex required for transport (ESCRT)-dependent micronuclear envelope repair by triggering autophagic degradation. We also found that p62 levels correlate with increased chromothripsis across human cancer cell lines and with increased CIN in colorectal tumors. Thus, p62 acts as a regulator of micronuclei and may serve as a prognostic marker for tumors with high CIN.


Subject(s)
Autophagy , Chromosomal Instability , Chromothripsis , Colorectal Neoplasms , Micronuclei, Chromosome-Defective , Sequestosome-1 Protein , Humans , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Cell Line, Tumor , DNA Damage , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Mitochondria/metabolism , Mitochondria/genetics , Nuclear Envelope/metabolism
15.
J Physiol ; 602(17): 4215-4235, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39167700

ABSTRACT

Oxidative stress contributes to the loss of skeletal muscle mass and function in cancer cachexia. However, this outcome may be mitigated by an improved endogenous antioxidant defence system. Here, using the well-established oxidative stress-inducing muscle atrophy model of Lewis lung carcinoma (LLC) in 13-week-old male C57BL/6J mice, we demonstrate that extracellular superoxide dismutase (EcSOD) levels increase in the cachexia-prone extensor digitorum longus muscle. LLC transplantation significantly increased interleukin-1ß (IL-1ß) expression and release from extensor digitorum longus muscle fibres. Moreover, IL-1ß treatment of C2C12 myotubes increased NBR1, p62 phosphorylation at Ser351, Nrf2 nuclear translocation and EcSOD protein expression. Additional studies in vivo indicated that intramuscular IL-1ß injection is sufficient to stimulate EcSOD expression, which is prevented by muscle-specific knockout of p62 and Nrf2 (i.e. in p62 skmKO and Nrf2 skmKO mice, respectively). Finally, since an increase in circulating IL-1ß may lead to unwanted outcomes, we demonstrate that targeting this pathway at p62 is sufficient to drive muscle EcSOD expression in an Nrf2-dependent manner. In summary, cancer cachexia increases EcSOD expression in extensor digitorum longus muscle via muscle-derived IL-1ß-induced upregulation of p62 phosphorylation and Nrf2 activation. These findings provide further mechanistic evidence for the therapeutic potential of p62 and Nrf2 to mitigate cancer cachexia-induced muscle atrophy. KEY POINTS: Oxidative stress plays an important role in muscle atrophy during cancer cachexia. EcSOD, which mitigates muscle loss during oxidative stress, is upregulated in 13-week-old male C57BL/6J mice of extensor digitorum longus muscles during cancer cachexia. Using mouse and cellular models, we demonstrate that cancer cachexia promotes muscle EcSOD protein expression via muscle-derived IL-1ß-dependent stimulation of the NBR1-p62-Nrf2 signalling pathway. These results provide further evidence for the potential therapeutic targeting of the NBR1-p62-Nrf2 signalling pathway downstream of IL-1ß to mitigate cancer cachexia-induced muscle atrophy.


Subject(s)
Cachexia , Interleukin-1beta , Mice, Inbred C57BL , Muscle, Skeletal , NF-E2-Related Factor 2 , Signal Transduction , Superoxide Dismutase , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Cachexia/metabolism , Cachexia/etiology , Cachexia/genetics , Male , Interleukin-1beta/metabolism , Muscle, Skeletal/metabolism , Mice , Superoxide Dismutase/metabolism , Superoxide Dismutase/genetics , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/complications , Carcinoma, Lewis Lung/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/genetics , Mice, Knockout , Oxidative Stress
16.
Neuromuscul Disord ; 42: 43-52, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39142003

ABSTRACT

TIA1/SQSTM1 myopathy is one of the few digenic myopathies. We describe four new French adult male patients carrying the TIA1 p.Asn357Ser and SQSTM1 p.Pro392Leu variant and review the literature to include 20 additional cases to define the spectrum of the disease. These twenty-four patients (75% males) had late-onset (52,6 ± 10,1 years), mainly asymmetric, distal ankle and hand finger extension weakness (75%), mild CK elevation (82.4%) and myopathic EMG. Two of the four French patients had sensorimotor axonal polyneuropathy and an additional one had neurogenic changes in muscle biopsy. Muscle biopsy showed rimmed vacuoles (44.4%), myofibrillar disorganization (16.7%) or both (38.9%), with P62/TDP43 aggregates. The TIA1 p.Asn357Ser variant was present in all patients and the SQSTM1 p.Pro392Leu was the most frequent (71%) of the four reported SQSTM1 variants. We reviewed the distal myopathy gene panels of Pitié-Salpêtrière's hospital cohort finding a prevalence of 11/414=2.7% of the TIA1 p.Asn357Ser variant, with two patients having an alternative diagnosis (TTN and MYH7) with atypical phenotypes, resembling some of the features seen in TIA1/SQSTM1 myopathy. Overall, TIA1/SQSTM1 myopathy has a homogenous phenotype reinforcing the pathogenicity of its digenic variants. We confirm an increased burden of the TIA1 p.Asn357Ser variant in distal myopathy patients which could act as a genetic modifier.


Subject(s)
Distal Myopathies , Sequestosome-1 Protein , T-Cell Intracellular Antigen-1 , Humans , Sequestosome-1 Protein/genetics , Male , Middle Aged , T-Cell Intracellular Antigen-1/genetics , Distal Myopathies/genetics , Distal Myopathies/pathology , Adult , Muscle, Skeletal/pathology , Aged , Female , Mutation , Phenotype
17.
Fish Shellfish Immunol ; 153: 109805, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39102972

ABSTRACT

The production of type I interferon is tightly regulated to prevent excessive immune activation. However, the role of selective autophagy receptor SQSTM1 in this regulation in teleost remains unknown. In this study, we cloned the triploid fish SQSTM1 (3nSQSTM1), which comprises 1371 nucleotides, encoding 457 amino acids. qRT-PCR data revealed that the transcript levels of SQSTM1 in triploid fish were increased both in vivo and in vitro following spring viraemia of carp virus (SVCV) infection. Immunofluorescence analysis confirmed that 3nSQSTM1 was mainly distributed in the cytoplasm. Luciferase reporter assay results showed that 3nSQSTM1 significantly blocked the activation of interferon promoters induced by 3nMDA5, 3nMAVS, 3nTBK1, and 3nIRF7. Co-immunoprecipitation assays further confirmed that 3nSQSTM1 could interact with both 3nTBK1 and 3nIRF7. Moreover, upon co-transfection, 3nSQSTM1 significantly inhibited the antiviral activity mediated by TBK1 and IRF7. Mechanistically, 3nSQSTM1 decreased the TBK1 phosphorylation and its interaction with 3nIRF7, thereby suppressing the subsequent antiviral response. Notably, we discovered that 3nSQSTM1 also interacted with SVCV N and P proteins, and these viral proteins may exploit 3nSQSTM1 to further limit the host's antiviral innate immune responses. In conclusion, our study demonstrates that 3nSQSTM1 plays a pivotal role in negatively regulating the interferon signaling pathway by targeting 3nTBK1 and 3nIRF7.


Subject(s)
Carps , Fish Diseases , Fish Proteins , Immunity, Innate , Interferon Regulatory Factor-7 , Rhabdoviridae Infections , Rhabdoviridae , Animals , Immunity, Innate/genetics , Fish Diseases/immunology , Fish Diseases/virology , Fish Proteins/genetics , Fish Proteins/immunology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Rhabdoviridae/physiology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/veterinary , Carps/immunology , Carps/genetics , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Gene Expression Regulation/immunology , Signal Transduction/immunology , Triploidy , Phylogeny , Amino Acid Sequence , Sequence Alignment/veterinary , Gene Expression Profiling/veterinary
18.
Free Radic Biol Med ; 222: 588-600, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996820

ABSTRACT

Emerging evidence has reported that acute lung injury (ALI), characterized by inflammation and oxidative stress in airway epithelium, is regulated by programmed cell death. Ferroptosis, a regulated form of cell death spurred by uncontrolled lipid peroxidation, has been proven to implicate various diseases. Inhibiting ferroptosis represents a feasible strategy for ALI through the suppression of lipid peroxidation, while the mechanism remains to be further elucidated. Here, we identified Sequestosome 1 (SQSTM1) as a negative regulator of airway epithelium ferroptosis during ALI. SQSTM1 knockdown cells manifested higher sensitivity to ferroptosis. Mechanistically, SQSTM1 was found to directly interact with vitamin D receptor (VDR) through its nuclear receptor (NR) box motif, facilitating its nuclear translocation and initiating autophagy at the transcriptional level. To further validate these findings, an in vivo preventive model utilizing spermidine, a proven inducer of SQSTM1 was established. The results consistently demonstrated that spermidine supplementation significantly induced SQSTM1 and ameliorated ALI by mitigating airway epithelial ferroptosis. Notably, these effects were abrogated in the absence of SQSTM1. Taken together, this study identified SQSTM1 as a negative regulator of airway epithelium ferroptosis in a VDR-mediated autophagy manner, making it a potential therapeutic target for the treatment of ALI.


Subject(s)
Acute Lung Injury , Autophagy , Ferroptosis , Receptors, Calcitriol , Sequestosome-1 Protein , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/genetics , Ferroptosis/genetics , Ferroptosis/drug effects , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/genetics , Animals , Humans , Mice , Male , Mice, Inbred C57BL , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Oxidative Stress , Lipid Peroxidation/drug effects
19.
Free Radic Biol Med ; 222: 607-624, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39009244

ABSTRACT

Autophagy is essential for the adaptive response to exercise and physiological skeletal muscle functionality. However, the mechanisms leading to the activation of macroautophagy and chaperone-mediated autophagy in human skeletal muscle in response to high-intensity exercise remain elusive. Our findings demonstrate that macroautophagy and chaperone-mediated autophagy are stimulated by high-intensity exercise in normoxia (PIO2: 143 mmHg) and severe acute hypoxia (PIO2: 73 mmHg) in healthy humans. High-intensity exercise induces macroautophagy initiation through AMPKα phosphorylation, which phosphorylates and activates ULK1. ULK1 phosphorylates BECN1 at Ser15, eliciting the dissociation of BECN1-BCL2 crucial for phagophore formation. Besides, high-intensity exercise elevates the LC3B-II:LC3B-I ratio, reduces total SQSTM1/p62 levels, and induces p-Ser349 SQSTM1/p62 phosphorylation, suggesting heightened autophagosome degradation. PHAF1/MYTHO, a novel macroautophagy biomarker, is highly upregulated in response to high-intensity exercise. The latter is accompanied by elevated LAMP2A expression, indicating chaperone-mediated autophagy activation regardless of post-exercise HSPA8/HSC70 downregulation. Despite increased glycolytic metabolism, severe acute hypoxia does not exacerbate the autophagy signaling response. Signaling changes revert within 1 min of recovery with free circulation, while the application of immediate post-exercise ischemia impedes recovery. Our study concludes that macroautophagy and chaperone-mediated autophagy pathways are strongly activated by high-intensity exercise, regardless of PO2, and that oxygenation is necessary to revert these signals to pre-exercise values. PHAF1/MYTHO emerges as a pivotal exercise-responsive autophagy marker positively associated with the LC3B-II:LC3B-I ratio.


Subject(s)
Autophagy-Related Protein-1 Homolog , Autophagy , Beclin-1 , Chaperone-Mediated Autophagy , Exercise , Hypoxia , Muscle, Skeletal , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Exercise/physiology , Male , Phosphorylation , Hypoxia/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Beclin-1/metabolism , Beclin-1/genetics , Chaperone-Mediated Autophagy/genetics , Ischemia/metabolism , Ischemia/pathology , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , AMP-Activated Protein Kinases/metabolism , Adult , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , HSC70 Heat-Shock Proteins/metabolism , HSC70 Heat-Shock Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Female
20.
J Agric Food Chem ; 72(30): 16998-17007, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39016055

ABSTRACT

Butachlor is widely used in agriculture around the world and therefore poses environmental and public health hazards due to persistent and poor biodegradability. Ferroptosis is a type of iron-mediated cell death controlled by glutathione (GSH) and GPX4 inhibition. P62 is an essential autophagy adaptor that regulates Keap1 to activate nuclear factor erythroid 2-related factor 2 (Nrf2), which effectively suppresses lipid peroxidation, thereby relieving ferroptosis. Here, we found that butachlor caused changes in splenic macrophage structure, especially impaired mitochondrial morphology with disordered structure, which is suggestive of the occurrence of ferroptosis. This was further confirmed by the detection of iron metabolism, the GSH system, and lipid peroxidation. Mechanistically, butachlor suppressed the protein level of p62 and promoted Keap1-mediated degradation of Nrf2, which results in decreased GPX4 expression and accelerated splenic macrophage ferroptosis. These findings suggest that targeting the p62-Nrf2-GPX4 signaling axis may be a promising strategy for treating inflammatory diseases.


Subject(s)
Ferroptosis , Macrophages , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Phospholipid Hydroperoxide Glutathione Peroxidase , Signal Transduction , Spleen , Animals , Humans , Male , Mice , Ferroptosis/drug effects , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Lipid Peroxidation/drug effects , Macrophages/drug effects , Macrophages/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Signal Transduction/drug effects , Spleen/drug effects , Spleen/cytology , Spleen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL