Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 990
Filter
1.
Cell Mol Biol Lett ; 29(1): 85, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834954

ABSTRACT

The molecular basis for bulk autophagy activation due to a deficiency in essential nutrients such as carbohydrates, amino acids, and nitrogen is well understood. Given autophagy functions to reduce surplus to compensate for scarcity, it theoretically possesses the capability to selectively degrade specific substrates to meet distinct metabolic demands. However, direct evidence is still lacking that substantiates the idea that autophagy selectively targets specific substrates (known as selective autophagy) to address particular nutritional needs. Recently, Gross et al. found that during phosphate starvation (P-S), rather than nitrogen starvation (N-S), yeasts selectively eliminate peroxisomes by dynamically altering the composition of the Atg1/ULK kinase complex (AKC) to adapt to P-S. This study elucidates how the metabolite sensor Pho81 flexibly interacts with AKC and guides selective autophagic clearance of peroxisomes during P-S, providing novel insights into the metabolic contribution of autophagy to special nutritional needs.


Subject(s)
Autophagy , Phosphates , Saccharomyces cerevisiae Proteins , Phosphates/metabolism , Phosphates/deficiency , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Peroxisomes/metabolism , Saccharomyces cerevisiae/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Kinases
2.
J Med Virol ; 96(6): e29690, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38804180

ABSTRACT

Autophagy is a degradational pathway with pivotal roles in cellular homeostasis and survival, including protection of neurons in the central nervous system (CNS). The significance of autophagy as antiviral defense mechanism is recognized and some viruses hijack and modulate this process to their advantage in certain cell types. Here, we present data demonstrating that the human neurotropic herpesvirus varicella zoster virus (VZV) induces autophagy in human SH-SY5Y neuronal cells, in which the pathway exerts antiviral activity. Productively VZV-infected SH-SY5Y cells showed increased LC3-I-LC3-II conversion as well as co-localization of the viral glycoprotein E and the autophagy receptor p62. The activation of autophagy was dependent on a functional viral genome. Interestingly, inducers of autophagy reduced viral transcription, whereas inhibition of autophagy increased viral transcript expression. Finally, the genotype of patients with severe ocular and brain VZV infection were analyzed to identify potential autophagy-associated inborn errors of immunity. Two patients expressing genetic variants in the autophagy genes ULK1 and MAP1LC3B2, respectively, were identified. Notably, cells of both patients showed reduced autophagy, alongside enhanced viral replication and death of VZV-infected cells. In conclusion, these results demonstrate a neuro-protective role for autophagy in the context of VZV infection and suggest that failure to mount an autophagy response is a potential predisposing factor for development of severe VZV disease.


Subject(s)
Autophagy , Herpesvirus 3, Human , Neurons , Humans , Herpesvirus 3, Human/physiology , Herpesvirus 3, Human/pathogenicity , Neurons/virology , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Virus Replication , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Varicella Zoster Virus Infection/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Cell Line , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Host-Pathogen Interactions
3.
BMC Pharmacol Toxicol ; 25(1): 33, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783387

ABSTRACT

BACKGROUND: The specific mechanism by which rotenone impacts thoracic aortic autophagy and apoptosis is unknown. We aimed to investigate the regulatory effects of rotenone on autophagy and apoptosis in rat thoracic aortic endothelial cells (RTAEC) via activation of the LKB1-AMPK-ULK1 signaling pathway and to elucidate the molecular mechanisms of rotenone on autophagy and apoptosis in vascular endothelial cells. METHODS: In vivo, 60 male SD rats were randomly selected and divided into 5 groups: control (Con), DMSO, 1, 2, and 4 mg/kg groups, respectively. After 28 days of treatment, histopathological and ultrastructural changes in each group were observed using HE and transmission electron microscopy; Autophagy, apoptosis, and LKB1-AMPK-ULK1 pathway-related proteins were detected by Western blot; Apoptosis levels in the thoracic aorta were detected by TUNEL. In vitro, RTAEC were cultured and divided into control (Con), DMSO, 20, 100, 500, and 1000 nM groups. After 24 h of intervention, autophagy, apoptosis, and LKB1-AMPK-ULK1 pathway-related factors were detected by Western blot and qRT-PCR; Flow cytometry to detect apoptosis levels; Autophagy was inhibited with 3-MA and CQ to detect apoptosis levels, and changes in autophagy, apoptosis, and downstream factors were detected by the AMPK inhibitor CC intervention. RESULTS: Gavage in SD rats for 28 days, some degree of damage was observed in the thoracic aorta and heart of the rotenone group, as well as the appearance of autophagic vesicles was observed in the thoracic aorta. TUNEL analysis revealed higher apoptosis in the rotenone group's thoracic aorta; RTAEC cultured in vitro, after 24 h of rotenone intervention, showed increased ROS production and significantly decreased ATP production. The flow cytometry data suggested an increase in the number of apoptotic RTAEC. The thoracic aorta and RTAEC in the rotenone group displayed elevated levels of autophagy and apoptosis, and the LKB1-AMPK-ULK1 pathway proteins were activated and expressed at higher levels. Apoptosis and autophagy were both suppressed by the autophagy inhibitors 3-MA and CQ. The AMPK inhibitor CC reduced autophagy and apoptosis in RTAEC and suppressed the production of the AMPK downstream factors ULK1 and P-ULK1. CONCLUSIONS: Rotenone may promote autophagy in the thoracic aorta and RTAEC by activating the LKB1-AMPK-ULK1 signaling pathway, thereby inducing apoptosis.


Subject(s)
AMP-Activated Protein Kinases , Aorta, Thoracic , Apoptosis , Autophagy-Related Protein-1 Homolog , Autophagy , Endothelial Cells , Protein Serine-Threonine Kinases , Rats, Sprague-Dawley , Rotenone , Signal Transduction , Animals , Rotenone/toxicity , Rotenone/pharmacology , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/metabolism , Male , Apoptosis/drug effects , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Aorta, Thoracic/drug effects , Protein Serine-Threonine Kinases/metabolism , Rats , AMP-Activated Protein Kinase Kinases , Cells, Cultured , Intracellular Signaling Peptides and Proteins/metabolism
4.
Theriogenology ; 225: 152-161, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38805997

ABSTRACT

Oocytes and embryos are highly sensitive to environmental stress in vivo and in vitro. During in vitro culture, many stressful conditions can affect embryo quality and viability, leading to adverse clinical outcomes such as abortion and congenital abnormalities. In this study, we found that valeric acid (VA) increased the mitochondrial membrane potential and ATP content, decreased the level of reactive oxygen species that the mitochondria generate, and thus improved mitochondrial function during early embryonic development in pigs. VA decreased expression of the autophagy-related factors LC3B and BECLIN1. Interestingly, VA inhibited expression of autophagy-associated phosphorylation-adenosine monophosphate-activated protein kinase (p-AMPK), phosphorylation-UNC-51-like autophagy-activated kinase 1 (p-ULK1, Ser555), and ATG13, which reduced apoptosis. Short-chain fatty acids (SCFAs) can signal through G-protein-coupled receptors on the cell membrane or enter the cell directly through transporters. We further show that the monocarboxylate transporter 1 (MCT1) was necessary for the effects of VA on embryo quality, which provides a new molecular perspective of the pathway by which SCFAs affect embryos. Importantly, VA significantly inhibited the AMPK-ULK1 autophagic signaling pathway through MCT1, decreased apoptosis, increased expression of embryonic pluripotency genes, and improved embryo quality.


Subject(s)
AMP-Activated Protein Kinases , Autophagy-Related Protein-1 Homolog , Autophagy , Embryonic Development , Mitochondria , Monocarboxylic Acid Transporters , Animals , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Swine/embryology , Embryonic Development/drug effects , Autophagy/drug effects , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/genetics , Signal Transduction/drug effects , Blastocyst/drug effects , Blastocyst/metabolism , Membrane Potential, Mitochondrial/drug effects , Embryo Culture Techniques/veterinary , Symporters
5.
Ecotoxicol Environ Saf ; 279: 116503, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38810288

ABSTRACT

Kashin-Beck disease (KBD) is an endemic, environmentally associated cartilage disease. Previous studies have shown that the environmental suspected pathogenic factors of KBD, T-2 toxin and low selenium, are involved in the regulation of inflammation, oxidative stress and autophagy in some tissues and organs. In cartilage diseases, the level of cellular autophagy determines the fate of the chondrocytes. However, whether autophagy is involved in KBD cartilage lesions, and the role of low selenium and T-2 toxins in KBD cartilage injury and autophagy are still unclear. This work took the classical AMPK/mTOR/ULK1 autophagy regulatory pathway as the entry point to clarify the relationship between the environmental suspected pathogenic factors and chondrocyte autophagy. Transmission electron microscopy was used to observe the autophagy of chondrocytes in KBD patients. qRT-PCR and western blot were used to analyze the expression of AMPK/mTOR/ULK1 pathway and autophagy markers. The rat model of KBD was established by low selenium and T-2 toxin, the autophagy in rat cartilage was detected after 4- and 12-week interventions. Chondrocyte autophagy was found in KBD, and the AMPK/mTOR/ULK1 pathway was down-regulated. In the rat model, the pathway showed an up-regulated trend when low selenium and T-2 toxin, were treated for a short time or low concentration, and autophagy level increased. However, when low selenium and T-2 toxin were treated for a long time or at high concentrations, the pathway showed a down-regulated trend, and the autophagy level was reduced and even defective. In conclusion, in the process of KBD cartilage lesion, chondrocyte autophagy level may increase in the early stage, and decrease in the late stage with the progression of lesion. Low selenium and T-2 toxins may affect autophagy by AMPK/mTOR/ULK1 pathway.


Subject(s)
AMP-Activated Protein Kinases , Autophagy-Related Protein-1 Homolog , Autophagy , Chondrocytes , Kashin-Beck Disease , Selenium , T-2 Toxin , TOR Serine-Threonine Kinases , T-2 Toxin/toxicity , T-2 Toxin/analogs & derivatives , Autophagy/drug effects , Kashin-Beck Disease/pathology , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy-Related Protein-1 Homolog/metabolism , Male , Chondrocytes/drug effects , Chondrocytes/pathology , Humans , AMP-Activated Protein Kinases/metabolism , Rats , Female , Middle Aged , Rats, Sprague-Dawley , Signal Transduction/drug effects , Adult , Intracellular Signaling Peptides and Proteins
6.
Int J Biol Sci ; 20(6): 2323-2338, 2024.
Article in English | MEDLINE | ID: mdl-38617533

ABSTRACT

Chronic atrophic gastritis (CAG) is a complex disease characterized by atrophy and inflammation in gastric mucosal tissue, especially with high expression of interleukins. However, the interaction and mechanisms between interleukins and gastric mucosal epithelial cells in CAG remain largely elusive. Here, we elucidate that IL-33 stands out as the predominant inflammatory factor in CAG, and its expression is induced by H. pylori and MNNG through the ROS-STAT3 signaling pathway. Furthermore, our findings reveal that the IL-33/ST2 axis is intricately involved in the progression of CAG. Utilizing phosphoproteomics mass spectrometry, we demonstrate that IL-33 enhances autophagy in gastric epithelial cells through the phosphorylation of AMPK-ULK1 axis. Notably, inhibiting autophagy alleviates CAG severity, while augmentation of autophagy exacerbates the disease. Additionally, ROS scavenging emerges as a promising strategy to ameliorate CAG by reducing IL-33 expression and inhibiting autophagy. Intriguingly, IL-33 stimulation promotes GKN1 degradation through the autolysosomal pathway. Clinically, the combined measurement of IL-33 and GKN1 in serum shows potential as diagnostic markers. Our findings unveil an IL-33-AMPK-ULK1 regulatory mechanism governing GKN1 protein stability in CAG, presenting potential therapeutic targets for its treatment.


Subject(s)
Gastritis, Atrophic , Helicobacter pylori , Peptide Hormones , Humans , AMP-Activated Protein Kinases , Autophagy-Related Protein-1 Homolog/genetics , Gastric Mucosa , Interleukin-33 , Intracellular Signaling Peptides and Proteins , Reactive Oxygen Species
7.
Am J Physiol Cell Physiol ; 326(6): C1590-C1603, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38586878

ABSTRACT

Muscular fatty infiltration is a common issue after rotator cuff tears (RCTs), which impair shoulder function. Females suffer a higher prevalence and a more severe degree of muscular fatty infiltration after RCT when compared with males, with the underlying mechanisms remaining unclear. Fibro-adipogenic progenitors (FAPs) are the primary source of muscular fatty infiltration following RCT. Our findings disclose that gender-specific disparities in muscular fatty infiltration are linked to mTOR/ULK1-mediated autophagy of FAPs. Decreased autophagic activity contributes to adipogenic differentiation in female FAPs after RCT. Furthermore, metformin could enhance mTOR/ULK1-mediated autophagic processes of FAPs, thereby alleviating fatty infiltration and improving shoulder functionality after RCT. Together, our study reveals that gender differences in muscular fatty infiltration arise from distinct autophagic activities. Metformin could be a promising noninvasive intervention to ameliorate muscular fatty infiltration of RCT.NEW & NOTEWORTHY The current study demonstrated that gender-specific disparities in muscular fatty infiltration are attributed to mTOR/ULK1-mediated autophagy of FAPs. Decreased autophagic activity contributes to adipogenic differentiation in female FAPs after RCT. Moreover, metformin could enhance mTOR/ULK1-mediated autophagic processes of FAPs, thereby alleviating fatty infiltration and improving shoulder functionality after RCT. Therefore, metformin could be a promising noninvasive intervention to ameliorate muscular fatty infiltration of RCT.


Subject(s)
Adipogenesis , Autophagy-Related Protein-1 Homolog , Autophagy , Metformin , Rotator Cuff Injuries , TOR Serine-Threonine Kinases , Animals , Autophagy/drug effects , Adipogenesis/drug effects , TOR Serine-Threonine Kinases/metabolism , Metformin/pharmacology , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Rotator Cuff Injuries/metabolism , Rotator Cuff Injuries/pathology , Rotator Cuff Injuries/drug therapy , Female , Male , Mice , Mice, Inbred C57BL , Stem Cells/drug effects , Stem Cells/metabolism , Cell Differentiation/drug effects , Signal Transduction/drug effects
8.
J Radiat Res ; 65(3): 291-302, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38588586

ABSTRACT

This study was aimed to investigate the effect of hydrogen-rich solution (HRS) on acute radiation pneumonitis (ARP) in rats. The ARP model was induced by X-ray irradiation. Histopathological changes were assessed using HE and Masson stains. Inflammatory cytokines were detected by ELISA. Immunohistochemistry and flow cytometry were performed to quantify macrophage (CD68) levels and the M2/M1 ratio. Western blot analysis, RT-qPCR, ELISA and flow cytometry were used to evaluate mitochondrial oxidative stress injury indicators. Immunofluorescence double staining was performed to colocalize CD68/LC3B and p-AMPK-α/CD68. The relative expression of proteins associated with autophagy activation and the adenosine 5'-monophosphate-activated protein kinase/mammalian target of rapamycin/Unc-51-like kinase 1 (AMPK/mTOR/ULK1) signaling pathway were detected by western blotting. ARP decreased body weight, increased the lung coefficient, collagen deposition and macrophage infiltration and promoted M1 polarization in rats. After HRS treatment, pathological damage was alleviated, and M1 polarization was inhibited. Furthermore, HRS treatment reversed the ARP-induced high levels of mitochondrial oxidative stress injury and autophagy inhibition. Importantly, the phosphorylation of AMPK-α was inhibited, the phosphorylation of mTOR and ULK1 was activated in ARP rats and this effect was reversed by HRS treatment. HRS inhibited M1 polarization and alleviated oxidative stress to activate autophagy in ARP rats by regulating the AMPK/mTOR/ULK1 signaling pathway.


Subject(s)
Autophagy , Hydrogen , Macrophages , Oxidative Stress , Radiation Pneumonitis , Rats, Sprague-Dawley , Animals , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Hydrogen/pharmacology , Hydrogen/therapeutic use , Autophagy/drug effects , Autophagy/radiation effects , Macrophages/drug effects , Macrophages/metabolism , Macrophages/radiation effects , Radiation Pneumonitis/drug therapy , Radiation Pneumonitis/pathology , Radiation Pneumonitis/metabolism , Male , Rats , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Cell Polarity/drug effects , Cell Polarity/radiation effects , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/radiation effects , Acute Disease
9.
Bioorg Chem ; 147: 107367, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38626492

ABSTRACT

Lung cancer is the leading cause of cancer deaths worldwide. Non-small cell lung cancer (NSCLC) accounts for 80-85% of all lung cancers. Euphorbia kansui yielded 13-oxyingenol-dodecanoate (13OD), an ingenane-type diterpenoid, which had a strong cytotoxic effect on NSCLC cells. The underlying mechanism and potential target, however, remained unknown. The study found that 13OD effectively inhibited the cell proliferation and colony formation of NSCLC cells (A549 and H460 cells), with less toxicity in normal human lung epithelial BEAS-2B cells. Moreover, 13OD can cause mitochondrial dysfunction, and apoptosis in NSCLC cells. Mechanistically, the transcriptomics results showed that differential genes were mainly enriched in the mTOR and AMPK signaling pathways, which are closely related to cellular autophagy, the related indicators were subsequently validated. Additionally, bafilomycin A1 (Baf A1), an autophagy inhibitor, reversed the mitochondrial damage caused by 13OD. Furthermore, the Omics and Text-based Target Enrichment and Ranking (OTTER) method predicted ULK1 as a potential target of 13OD against NSCLC cells. This hypothesis was further confirmed using molecular docking, the cellular thermal shift assay (CETSA), and Western blot analysis. Remarkably, ULK1 siRNA inhibited 13OD's toxic activity in NSCLC cells. In line with these findings, 13OD was potent and non-toxic in the tumor xenograft model. Our findings suggested a possible mechanism for 13OD's role as a tumor suppressor and laid the groundwork for identifying targets for ingenane-type diterpenoids.


Subject(s)
Autophagy-Related Protein-1 Homolog , Carcinoma, Non-Small-Cell Lung , Cell Proliferation , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Structure-Activity Relationship , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Molecular Structure , Diterpenes/pharmacology , Diterpenes/chemistry , Apoptosis/drug effects , Animals , Mice , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis
10.
Zhongguo Zhong Yao Za Zhi ; 49(5): 1286-1294, 2024 Mar.
Article in Chinese | MEDLINE | ID: mdl-38621976

ABSTRACT

This study explored the specific mechanism by which tetrahydropalmatine(THP) inhibited mitophagy through the UNC-51-like kinase 1(ULK1)/FUN14 domain containing 1(FUNDC1) pathway to reduce hypoxia/reoxygenation(H/R) injury in H9c2 cells. This study used H9c2 cells as the research object to construct a cardiomyocyte H/R injury model. First, a cell viability detection kit was used to detect cell viability, and a micro-method was used to detect lactate dehydrogenase(LDH) leakage to evaluate the protective effect of THP on H/R injury of H9c2 cells. In order to evaluate the protective effect of THP on mitochondria, the chemical fluorescence method was used to detect intracellular reactive oxygen species, intramitochondrial reactive oxygen species, mitochondrial membrane potential, and autophagosomes, and the luciferin method was used to detect intracellular adenosine 5'-triphosphate(ATP) content. Western blot was further used to detect the ratio of microtubule-associated protein 1 light chain 3(LC3) membrane type(LC3-Ⅱ) and slurry type(LC3-Ⅰ) and activated cleaved caspase-3 expression level. In addition, ULK1 expression level and its phosphorylation degree at Ser555 site, as well as the FUNDC1 expression level and its phosphorylation degree of Ser17 site were detected to explore its specific mechanism. The results showed that THP effectively reduced mitochondrial damage in H9c2 cells after H/R. THP protected mitochondria by reducing the level of reactive oxygen species in cells and mitochondria, increasing mitochondrial membrane potential, thereby increasing cellular ATP production, enhancing cellular activity, reducing cellular LDH leakage, and finally alleviating H/R damage in H9c2 cells. Further studies have found that THP could reduce the production of autophagosomes, reduce the LC3-Ⅱ/LC3-Ⅰ ratio, and lower the expression of the apoptosis-related protein, namely cleaved caspase-3, indicating that THP could reduce apoptosis by inhibiting autophagy. In-depth studies have found that THP could inhibit the activation of the ULK1/FUNDC1 pathway of mitophagy and the occurrence of mitophagy by reducing the phosphorylation degree of ULK1 at Ser555 and FUNDC1 at Ser17. The application of ULK1 agonist BL-918 reversely verified the effect of THP on reducing the phosphorylation of ULK1 and FUNDC1. In summary, THP inhibited mitophagy through the ULK1/FUNDC1 pathway to reduce H/R injury in H9c2 cells.


Subject(s)
Berberine Alkaloids , Hypoxia , Mitophagy , Phenylacetates , Humans , Mitophagy/physiology , Caspase 3 , Reactive Oxygen Species/metabolism , Apoptosis , Adenosine Triphosphate/pharmacology , Autophagy-Related Protein-1 Homolog/genetics , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mitochondrial Proteins
11.
Cancer Biol Ther ; 25(1): 2334463, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38569536

ABSTRACT

Neurensin-2 (NRSN2) performs a pro-carcinogenic function in multiple cancers. However, the function of NRSN2 in HPV-infected laryngeal carcinoma (LC) remains unclear. HPV transfection was performed in LC cells. The mRNA and protein levels were monitored using RT-qPCR, immunoblotting, and IF. Cell viability and proliferation were found using the CCK-8 assay and Edu staining. Cell invasion, migration, and apoptosis were probed using the Transwell, wound healing, and flow cytometry, respectively. The autophagosome was observed using TEM. NRSN2 was overexpressed in HPV-transfected LC cells. Inhibition of NRSN2 restrained the autophagy and malignant behavior of HPV-transfected LC cells. Meanwhile, the inhibition of AMPK/ULK1 pathway limited the increased autophagy of HPV-transfected LC cells caused by NRSN2 overexpression. Furthermore, NRSN2 knockdown inhibits autophagy by suppressing AMPK/ULK1 pathway, thereby restraining the malignant behavior of HPV-transfected LC cells. Our research confirmed that HPV transfection increased the autophagy and malignant behavior of LC cells by regulating the NRSN2-mediated activation of the AMPK/ULK1 pathway, offering a new target for cure of LC.


Subject(s)
Carcinoma , Papillomavirus Infections , Humans , AMP-Activated Protein Kinases , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy/genetics , Intracellular Signaling Peptides and Proteins
12.
Trends Biochem Sci ; 49(6): 494-505, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38565496

ABSTRACT

Autophagy - a highly regulated intracellular degradation process - is pivotal in maintaining cellular homeostasis. Liquid-liquid phase separation (LLPS) is a fundamental mechanism regulating the formation and function of membrane-less compartments. Recent research has unveiled connections between LLPS and autophagy, suggesting that phase separation events may orchestrate the spatiotemporal organization of autophagic machinery and cargo sequestration. The Unc-51-like kinase (ULK)/autophagy-related 1 (Atg1) family of proteins is best known for its regulatory role in initiating autophagy, but there is growing evidence that the functional spectrum of ULK/Atg1 extends beyond autophagy regulation. In this review, we explore the spatial and temporal regulation of the ULK/Atg1 family of kinases, focusing on their recruitment to LLPS-driven compartments, and highlighting their multifaceted functions beyond their traditional role.


Subject(s)
Autophagy-Related Protein-1 Homolog , Autophagy , Humans , Autophagy-Related Protein-1 Homolog/metabolism , Animals , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/chemistry
13.
Oncogene ; 43(23): 1769-1778, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38632437

ABSTRACT

Pyruvate kinase M2 (PKM2) is a central metabolic enzyme driving the Warburg effect in tumor growth. Previous investigations have demonstrated that PKM2 is subject to O-linked ß-N-acetylglucosamine (O-GlcNAc) modification, which is a nutrient-sensitive post-translational modification. Here we found that unc-51 like autophagy activating kinase 1 (ULK1), a glucose-sensitive kinase, interacts with PKM2 and phosphorylates PKM2 at Ser333. Ser333 phosphorylation antagonizes PKM2 O-GlcNAcylation, promotes its tetramer formation and enzymatic activity, and decreases its nuclear localization. As PKM2 is known to have a nuclear role in regulating c-Myc, we also show that PKM2-S333 phosphorylation inhibits c-Myc expression. By downregulating glucose consumption and lactate production, PKM2 pS333 attenuates the Warburg effect. Through mouse xenograft assays, we demonstrate that the phospho-deficient PKM2-S333A mutant promotes tumor growth in vivo. In conclusion, we identified a ULK1-PKM2-c-Myc axis in inhibiting breast cancer, and a glucose-sensitive phosphorylation of PKM2 in modulating the Warburg effect.


Subject(s)
Autophagy-Related Protein-1 Homolog , Breast Neoplasms , Carrier Proteins , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Warburg Effect, Oncologic , Humans , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Phosphorylation , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Female , Mice , Thyroid Hormones/metabolism , Thyroid Hormones/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Cell Line, Tumor , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Acetylglucosamine/metabolism
14.
Acta Biochim Biophys Sin (Shanghai) ; 56(5): 789-804, 2024 05 25.
Article in English | MEDLINE | ID: mdl-38686458

ABSTRACT

Acute lung injury (ALI) is a serious condition characterized by damage to the lungs. Recent research has revealed that activation of the NLRP3 inflammasome in alveolar macrophages, a type of immune cell in the lungs, plays a key role in the development of ALI. This process, known as pyroptosis, contributes significantly to ALI pathogenesis. Researchers have conducted comprehensive bioinformatics analyses and identified 15 key genes associated with alveolar macrophage pyroptosis in ALI. Among these, NLRP3 has emerged as a crucial regulator. This study further reveal that the ULK1 protein diminishes the expression of NLRP3, thereby reducing the immune response of alveolar macrophages and mitigating ALI. Conversely, TRAF3, another protein, is found to inhibit ULK1 through a process called ubiquitination, leading to increased activation of the NLRP3 inflammasome and exacerbation of ALI. This TRAF3-mediated suppression of ULK1 and subsequent activation of NLRP3 are confirmed through various in vitro and in vivo experiments. The presence of abundant M0 and M1 alveolar macrophages in the ALI tissue samples further support these findings. This research highlights the TRAF3-ULK1-NLRP3 regulatory axis as a pivotal pathway in ALI development and suggests that targeting this axis could be an effective therapeutic strategy for ALI treatment.


Subject(s)
Acute Lung Injury , Autophagy-Related Protein-1 Homolog , Macrophages, Alveolar , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , TNF Receptor-Associated Factor 3 , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Animals , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Mice , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 3/genetics , Humans , Male , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Signal Transduction , Ubiquitination
15.
Cell Biol Int ; 48(6): 821-834, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38436129

ABSTRACT

Keratinocytes, located in the outermost layer of human skin, are pivotal cells to resist environmental damage. Cellular autophagy plays a critical role in eliminating damaged organelles and maintaining skin cell homeostasis. Low-dose 5-Aminolevulinic acid photodynamic therapy (ALA-PDT) has been demonstrated to enhance skin's antistress ability; however, the regulatory mechanisms of autophagy in keratinocytes remain unclear. In this study, we treated immortalized human keratinocytes (HaCaT cells) with low-dose ALA-PDT (0.5 mmol/L, 3 J/cm2). Through RNA-sequencing analysis, we identified that low-dose ALA-PDT modulated autophagy-related pathways in keratinocytes and pinpointed Unc-51-like kinase 1 (ULK1) as a key gene involved. Western blot results revealed that low-dose ALA-PDT treatment upregulated the expression of autophagy-related proteins Beclin-1 and LC3-II/LC3-I ratio. Notably, low-dose ALA-PDT regulated autophagy by inducing an appropriate level of reactive oxygen species (ROS), transiently reducing mitochondrial membrane potential, and decreasing adenosine triphosphate production; all these processes functioned on the AMP-activated protein kinase (AMPK)/ULK1 pathway to activate autophagy. Finally, we simulated external environmental damage using ultraviolet B (UVB) at a dose of 60 mJ/cm2 and observed that low-dose ALA-PDT mitigated UVB-induced cell apoptosis; however, this protective effect was reversed when using the autophagy inhibitor 3-methyladenine. Overall, these findings highlight how low-dose ALA-PDT enhances antistress ability in HaCaT cells through controlling ROS generation and activating the AMPK/ULK1 pathway to arouse cellular autophagy.


Subject(s)
AMP-Activated Protein Kinases , Autophagy-Related Protein-1 Homolog , Autophagy , Keratinocytes , Signal Transduction , Humans , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy/drug effects , Keratinocytes/metabolism , Keratinocytes/drug effects , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Aminolevulinic Acid/pharmacology , HaCaT Cells , Membrane Potential, Mitochondrial/drug effects
16.
J Nutr Biochem ; 129: 109623, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38492819

ABSTRACT

Chemotherapy failure in colorectal cancer patients is the major cause of recurrence and poor prognosis. As a result, there is an urgent need to develop drugs that have a good chemotherapy effect while also being extremely safe. In this study, we found cafestol inhibited colon cancer growth and HCT116 proliferation in vivo and in vitro, and improved the composition of intestinal flora. Further metabolomic data showed that autophagy and AMPK pathways were involved in the process of cafestol's anti-colon cancer effects. The functional validation studies revealed that cafestol increased autophagy vesicles and LC3B-II levels. The autophagic flux induced by cafestol was prevented by using BafA1. The autophagy inhibitor 3-MA blocked the cafestol-induced increase in LC3B-II and cell proliferation inhibition. Then we found that cafestol induced the increased expressions of LKB1, AMPK, ULK1, p-LKB1, p-AMPK, and p-ULK1 proteins in vivo and in vitro. Using the siRNA targeted to the Lkb1 gene, the levels of AMPK, ULK1, and LC3B-II were suppressed under cafestol treatment. These results indicated that the effect of cafestol is through regulating LKB1/AMPK/ULK1 pathway-mediated autophagic death. Finally, a correlation matrix of the microbiome and autophagy-related proteins was conducted. We found that cafestol-induced autophagic protein expression was positively correlated with the beneficial intestinal bacteria (Muribaculaceae, Bacteroides, Prevotellacece, and Alloprevotella) and negatively correlated with the hazardous bacteria. Conclusions: This study found that cafestol inhibited colon cancer in vitro and in vivo by the mechanism that may be related to LKB1/AMPK/ULK1 pathway-mediated autophagic cell death and improved intestinal microenvironment.


Subject(s)
AMP-Activated Protein Kinases , Autophagy-Related Protein-1 Homolog , Autophagy , Cell Proliferation , Colonic Neoplasms , Protein Serine-Threonine Kinases , Xenograft Model Antitumor Assays , Animals , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy/drug effects , Humans , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cell Proliferation/drug effects , AMP-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Mice , HCT116 Cells , AMP-Activated Protein Kinase Kinases , Mice, Nude , Mice, Inbred BALB C , Gastrointestinal Microbiome/drug effects , Signal Transduction/drug effects , Male
17.
Adv Sci (Weinh) ; 11(18): e2310065, 2024 May.
Article in English | MEDLINE | ID: mdl-38447147

ABSTRACT

According to the latest evidence, the microbial metabolite Urolithin A (UA), known for its role in promoting cellular health, modulates CD8+ T cell-mediated antitumor activity. However, the direct target protein of UA and its underlying mechanism remains unclear. Here, this research identifies ERK1/2 as the specific target crucial for UA-mediated CD8+ T cell activation. Even at low doses, UA markedly enhances the persistence and effector functions of primary CD8+ cytotoxic T lymphocytes (CTLs) and human chimeric antigen receptor (CAR) T cells both in vitro and in vivo. Mechanistically, UA interacts directly with ERK1/2 kinases, enhancing their activation and subsequently facilitating T cell activation by engaging ULK1. The UA-ERK1/2-ULK1 axis promotes autophagic flux in CD8+ CTLs, enhancing cellular metabolism and maintaining reactive oxygen species (ROS) levels, as evidenced by increased oxygen consumption and extracellular acidification rates. UA-treated CD8+ CTLs also display elevated ATP levels and enhanced spare respiratory capacity. Overall, UA activates ERK1/2, inducing autophagy and metabolic adaptation, showcasing its potential in tumor immunotherapy and interventions for diseases involving ERKs.


Subject(s)
Autophagy-Related Protein-1 Homolog , CD8-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Animals , Mice , Humans , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , MAP Kinase Signaling System/immunology , Coumarins/pharmacology , Coumarins/metabolism , Disease Models, Animal , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Mice, Inbred C57BL , Autophagy/immunology
18.
Phytomedicine ; 128: 155512, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38460357

ABSTRACT

BACKGROUND: The overproliferation of fibroblast-like synoviocytes (FLS) contributes to synovial hyperplasia, a pivotal pathological feature of rheumatoid arthritis (RA). Shikonin (SKN), the active compound from Lithospermum erythrorhizon, exerts anti-RA effects by diverse means. However, further research is needed to confirm SKN's in vitro and in vivo anti-proliferative functions and reveal the underlying specific molecular mechanisms. PURPOSE: This study revealed SKN's anti-proliferative effects by inducing both apoptosis and autophagic cell death in RA FLS and adjuvant-induced arthritis (AIA) rat synovium, with involvement of regulating the AMPK/mTOR/ULK-1 pathway. METHODS: SKN's influences on RA FLS were assessed for proliferation, apoptosis, and autophagy with immunofluorescence staining (Ki67, LC3B, P62), EdU incorporation assay, staining assays of Hoechst, Annexin V-FITC/PI, and JC-1, transmission electron microscopy, mCherry-GFP-LC3B puncta assay, and western blot. In AIA rats, SKN's anti-arthritic effects were assessed, and its impacts on synovial proliferation, apoptosis, and autophagy were studied using Ki67 immunohistochemistry, TUNEL, and western blot. The involvement of AMPK/mTOR/ULK-1 pathway was examined via western blot. RESULTS: SKN suppressed RA FLS proliferation with reduced cell viability and decreased Ki67-positive and EdU-positive cells. SKN promoted RA FLS apoptosis, as evidenced by apoptotic nuclear fragmentation, increased Annexin V-FITC/PI-stained cells, reduced mitochondrial potential, elevated Bax/Bcl-2 ratio, and increased cleaved-caspase 3 and cleaved-PARP protein levels. SKN also enhanced RA FLS autophagy, featuring increased LC3B, reduced P62, autophagosome formation, and activated autophagic flux. Autophagy inhibition by 3-MA attenuated SKN's anti-proliferative roles, implying that SKN-induced autophagy contributes to cell death. In vivo, SKN mitigated the severity of rat AIA while also reducing Ki67 expression, inducing apoptosis, and enhancing autophagy within AIA rat synovium. Mechanistically, SKN modulated the AMPK/mTOR/ULK-1 pathway in RA FLS and AIA rat synovium, as shown by elevated P-AMPK and P-ULK-1 expression and decreased P-mTOR expression. This regulation was supported by the reversal of SKN's in vitro and in vivo effects upon co-administration with the AMPK inhibitor compound C. CONCLUSION: SKN exerted in vitro and in vivo anti-proliferative properties by inducing apoptosis and autophagic cell death via modulating the AMPK/mTOR/ULK-1 pathway. Our study revealed novel molecular mechanisms underlying SKN's anti-RA effects.


Subject(s)
AMP-Activated Protein Kinases , Apoptosis , Arthritis, Experimental , Arthritis, Rheumatoid , Autophagy-Related Protein-1 Homolog , Autophagy , Naphthoquinones , Signal Transduction , Synoviocytes , TOR Serine-Threonine Kinases , Animals , TOR Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Arthritis, Rheumatoid/drug therapy , Naphthoquinones/pharmacology , Signal Transduction/drug effects , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/metabolism , AMP-Activated Protein Kinases/metabolism , Rats , Arthritis, Experimental/drug therapy , Synoviocytes/drug effects , Synoviocytes/metabolism , Male , Cell Proliferation/drug effects , Humans , Rats, Sprague-Dawley
19.
Allergol Immunopathol (Madr) ; 52(2): 3-9, 2024.
Article in English | MEDLINE | ID: mdl-38459884

ABSTRACT

OBJECTIVE: To detect the expression of RING finger protein 135 (RNF135) in lung adenocarcinoma tissues and explore its role in the progression of lung adenocarcinoma. METHODS: Bioinformation analysis, quantitative polymerase chain reaction, and immunoblotting technique discovered the expression of RNF135 in lung adenocarcinoma tissues. Cell counting kit-8 and colony formation, immunostaining, and immunoblot assays examined the effects of RNF135 on cell growth and autophagy. Co-immunoprecipitation (Co-IP), immunostaining, and immuoblotting were conducted to confirm the mechanism. RESULTS: RNF135 was highly expressed in lung adenocarcinoma. In addition, RNF135 promoted lung adenocarcinoma cell growth. Further, data confirmed that RNF135 promoted autophagy in lung adenocarcinoma cells. Mechanically, RNF135 directly interacted with Unc-51-like autophagy activating kinase 1 (ULK1) to promote its phosphorylation level. CONCLUSION: RNF135 promoted cell growth and autophagy in lung adenocarcinoma by promoting the phosphorylation of ULK1.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Ubiquitin-Protein Ligases , Humans , Autophagy , Autophagy-Related Protein-1 Homolog/metabolism , Cell Proliferation , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Lung Neoplasms/pathology , Phosphorylation , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/pharmacology
20.
Eur J Med Chem ; 268: 116273, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38432059

ABSTRACT

Autophagy is a process of self-renewal in cells, which not only provides the necessary nutrients for cells, but also clears necrotic organelles. Autophagy disorders are closely related to diseases such as cancer. UNC-51-like kinase 1 (ULK1) is a serine/threonine protein kinase that plays a crucial role in receiving input from energy and nutrient sensors, activating autophagy to maintain cellular homeostasis under stressful conditions. In recent years, targeting ULK1 has become a highly promising strategy for cancer treatment. This review introduces the regulatory mechanism of ULK1 in autophagy through the AMPK/mTOR/ULK1 pathway and reviews the research progress of ULK1 activators and inhibitors and their applications in cancer treatment. In addition, we analyze the binding modes between ULK1 and modulators through virtual molecular docking, which will provide a reliable basis and theoretical guidance for the design and development of new therapeutic drugs targeting ULK1.


Subject(s)
AMP-Activated Protein Kinases , Neoplasms , Autophagy-Related Protein-1 Homolog/metabolism , Molecular Docking Simulation , AMP-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Autophagy , Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...